US11065278B2 - Method and compositions for cellular immunotherapy - Google Patents

Method and compositions for cellular immunotherapy Download PDF

Info

Publication number
US11065278B2
US11065278B2 US15/969,438 US201815969438A US11065278B2 US 11065278 B2 US11065278 B2 US 11065278B2 US 201815969438 A US201815969438 A US 201815969438A US 11065278 B2 US11065278 B2 US 11065278B2
Authority
US
United States
Prior art keywords
cells
cell
car
lymphocytes
ror1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US15/969,438
Other versions
US20180296602A1 (en
Inventor
Stanley R. Riddell
Michael Hudecek
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fred Hutchinson Cancer Center
Original Assignee
Fred Hutchinson Cancer Research Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46208746&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US11065278(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Fred Hutchinson Cancer Research Center filed Critical Fred Hutchinson Cancer Research Center
Priority to US15/969,438 priority Critical patent/US11065278B2/en
Publication of US20180296602A1 publication Critical patent/US20180296602A1/en
Application granted granted Critical
Publication of US11065278B2 publication Critical patent/US11065278B2/en
Assigned to FRED HUTCHINSON CANCER CENTER reassignment FRED HUTCHINSON CANCER CENTER MERGER AND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: FRED HUTCHINSON CANCER RESEARCH CENTER, SEATTLE CANCER CARE ALLIANCE
Assigned to FRED HUTCHINSON CANCER CENTER reassignment FRED HUTCHINSON CANCER CENTER MERGER AND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: FRED HUTCHINSON CANCER RESEARCH CENTER, SEATTLE CANCER CARE ALLIANCE
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464424CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464838Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to the field of biomedicine and specifically methods useful for cancer therapy.
  • embodiments of the invention relate to methods and compositions for carrying out cellular immunotherapy.
  • T M The pool of lymphocytes from which T cells for adoptive immunotherapy can be derived contains na ⁇ ve and long-lived, antigen experienced memory T cells (T M ).
  • T M can be divided further into subsets of central memory (T CM ) and effector memory (T EM ) cells that differ in phenotype, homing properties and function 18 .
  • CD8 + T CM express CD62L and CCR7 at the cell surface, which promote migration into lymph nodes, and proliferate rapidly if re-exposed to antigen.
  • CD8 + T EM lack cell surface CD62L and preferentially migrate to peripheral tissues, and exhibit immediate effector function 19 .
  • T E cytolytic effector T cells
  • CD8 + T CM and T EM both differentiate into cytolytic effector T cells (T E ) that express a high level of granzymes and perforin, but are short-lived 20 .
  • T E cytolytic effector T cells
  • the poor survival of T cells in clinical immunotherapy trials may simply result from their differentiation during in vitro culture to T E that are destined to die 17,21,22 .
  • the present invention relates to methods and compositions to confer and/or augment immune responses mediated by cellular immunotherapy, such as by adoptively transferring tumor-specific, subset specific genetically modified CD4+ T cells, wherein the CD4+ T cells confer and/or augment the ability of CD8+ T cells to sustain anti-tumor reactivity and increase and/or maximize tumor-specific proliferation.
  • the present invention provides a method of performing cellular immunotherapy in a subject having a disease or disorder by administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation that provides a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as co-stimulatory domains; and a genetically modified helper T lymphocyte cell preparation that exhibits a predominant Th1 phenotype as well as produce other cytokines, elicits direct tumor recognition and augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor
  • the chimeric antigen receptor modifying the CD4+ T cell and the CD8+ T cell can be the same or different.
  • the T cells can be modified with a recombinant T cell receptor (TCR).
  • TCR could be specific for any antigen, pathogen or tumor.
  • TCRs for many tumor antigens in melanoma MART1, gp100, for example
  • WT1 leukemia
  • minor histocompatibility antigens for example
  • breast cancer here2, NY-BR1, for example.
  • the present invention provides an adoptive cellular immunotherapy composition having a genetically modified CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response
  • the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular variable domain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as a costimulatory domain, and a genetically modified helper T lymphocyte cell preparation that exhibits a predominant Th1 phenotype as well as produce other cytokines, elicits direct tumor recognition and augments the ability of genetically modified cytotoxic T lymphocyte cell preparations to mediate a cellular immune response
  • the helper T lymphocyte cell preparation has CD4+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • the present invention provides an adoptive cellular immunotherapy composition having a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and an antigen-reactive chimeric antigen receptor modified na ⁇ ve CD4+ T helper cell that is derived from CD45RO negative, CD62L positive CD4 positive T cells, and a pharmaceutically acceptable carrier.
  • the present invention provides an adoptive cellular immunotherapy composition having an antigen specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response comprising CD8+ T cells derived from the patient together with an antigen-reactive chimeric antigen receptor modified CD4+ T helper cell that elicits a Th1 cytokine response and augments the CD8+ immune response to pathogens, wherein the helper T lymphocyte cell preparation with CD4+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • the present invention provides an adoptive cellular immunotherapy composition with an antigen-reactive chimeric antigen receptor modified CD4+ T helper cell that elicits direct tumor recognition and augments the CD8+ immune response to pathogens, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with a disease or disorder and an intracellular signaling domain of a T cell receptor.
  • the present invention provides a method of manufacturing an adoptive immunotherapy composition by obtaining a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response and an antigen-reactive chimeric antigen receptor, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor; and obtaining a modified na ⁇ ve CD4+ T helper cell that elicits a Th1 cytokine response, wherein the modified helper T lymphocyte cell preparation comprises CD4+ cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • the present invention provides a method of manufacturing an adoptive immunotherapy composition by obtaining a modified na ⁇ ve CD4+ T helper cell that elicits a Th1 cytokine response, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and combining the modified na ⁇ ve CD4+ T helper cell with an antigen specific central memory CD8+ cytotoxic T lymphocyte cell preparation that has a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors.
  • the present invention provides a method of performing cellular immunotherapy in subject having a disease or disorder by administering to the subject a genetically modified helper T lymphocyte cell preparation, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor.
  • FIG. 1 shows the phenotype and analysis of chimeric antigen receptor (CAR) expression in a CAR-transduced with ROR1-CAR encoding lentivirus, and an untransduced CD8+ T cell line as a control.
  • the ROR1-CAR cassette contains a truncated EGFR that serves as transduction marker and can be detected by staining with anti-EGFR monoclonal antibodies.
  • Truncated Fc-ROR1 fusion protein binds directly to the antigen-binding domain of the ROR1-CAR and selectively stains the ROR1-CAR transduced but not the untransduced control T cell line.
  • ROR1-CAR expression of the ROR1-CAR on the cell surface of CD8+ T cells is measured directly by binding to ROR1-Fc fusion protein and indirectly by expression of a truncated EGFR that is encoded downstream of a 2A sequence in the vector.
  • FIG. 2 shows cytolytic activity of CD8+ T cells expressing a ROR1-specific chimeric antigen receptor against a panel of human ROR1-positive tumor cell lines (K562) and primary tumor cells (B-CLL) and autologous normal B-cells in a 51 Cr release assay. Consistent with the uniform expression of ROR1 on malignant but not on mature normal B cells, genetically modified CD8+ ROR1-CAR T cells only lysed ROR1+ tumor cells but not mature normal B cells. CD8+ ROR1-CAR T cells exert specific lytic activity against ROR1-positive tumor cells including primary CLL, but not against normal B cells.
  • FIG. 3 shows the phenotype and CAR expression of a ROR1-CAR transduced and an untransduced CD4+ T cell line as a control.
  • Expression of the ROR1-CAR on the cell surface of CD4+ T cells is measured by specific binding to ROR1-Fc fusion protein.
  • Truncated Fc ROR1 fusion protein but not Fc protein alone binds directly to the ROR1-CAR and selectively stains the ROR1-CAR transduced but not the untransduced control CD4+ T cell line confirming expression of the ROR1-CAR on the cell surface and binding to ROR1-protein.
  • Expression of the ROR1-CAR on the cell surface of CD4+ T cells is measured by specific binding to ROR1-Fc fusion protein, but not to a control Fc fusion protein.
  • FIGS. 4A and 4B show weak but specific cytolytic activity of CD4+ ROR1-CAR T cells in a 51 Cr release assay against a panel of ROR1-positive tumor cells including primary CLL, the mantle cell lymphoma line Jeko-1, K562 cells that were stably transfected with ROR1 (K562/ROR1), but not native ROR1-negative K562 cells.
  • CD4+ ROR1-CAR T cells exert weak but specific lytic activity against ROR1-positive tumor cells.
  • FIGS. 5A and 5B show the results from an IFN ⁇ ELISA ( FIG. 5A ) and multiplex cytokine assay ( FIG. 5B ).
  • Cytokine secretion of CD4+ and CD8+ ROR1-CAR T cell lines CD4+ROR1-CAR and CD8 ROR1-CAR T cells were co-incubated with ROR1+ tumor cells, and levels of interferon gamma (IFN ⁇ ) was measured by ELISA ( 5 A), and IFN ⁇ , TNF ⁇ , IL-2, IL-4, IL-10 and IL-17 were measured by Luminex assay ( 5 B).
  • IFN ⁇ interferon gamma
  • CD4+ ROR1-CAR modified T cells specifically recognize ROR1-positive tumor cells and tumor cell lines and produce higher amounts of Th1 cytokines including IFN- ⁇ , TNF- ⁇ and particularly IL-2 than CD8+ ROR1-CAR modified T cells.
  • FIG. 6 depicts the results of a proliferation study showing that CD4+ ROR1-CAR T cells are induced to proliferate after stimulation with ROR1-positive tumor cell lines and primary tumor cells (CFSE assay) and that both the percentage of proliferating cells and number of cell divisions that the proliferating subset underwent were significantly higher compared to CD8+ ROR1-CAR modified T cells.
  • CD4+ ROR1-CAR T cells proliferate more vigorously after stimulation with ROR1-positive tumor cells (K562/ROR1, primary CLL, and Jeko MCL) compared to CD8+ ROR1-CAR CTLs.
  • FIG. 7 shows polyclonal unselected CD4+ ROR1 CAR T cells provide help to CD8+ ROR1-CAR CTLs by promoting their proliferation in response to tumor.
  • CD4+ ROR1-CAR T cells derived from bulk CD4+ T cells
  • FIGS. 8A-8D show the generation of CD4+ CAR T cell lines from flow sort purified CD4+ na ⁇ ve, central memory and effector memory subsets and analysis of T-cell function. Cytokine profile and proliferative capacity suggest that CD4+ ROR1-CAR T cells derived from na ⁇ ve CD4+ T cells may be best suited to provide help to CD8+ CTLs. Similar data were obtained in experiments comparing the function of CD4+ CAR T-cell lines expressing a CD19-specific CAR.
  • FIG. 8A shows flow sort purification of na ⁇ ve, central and effector memory CD4+ T cells based on expression of CD45RA, CD45RO, CD62L. FIG.
  • FIG. 8B shows analysis of proliferation of ROR1-CAR T cell lines that were derived by lentiviral transduction of sort purified na ⁇ ve, central and effector memory CD4+ T cells (CFSE assay).
  • FIG. 8C shows analysis of cytokine secretion of ROR1-CAR T cell lines from sort purified na ⁇ ve, central and effector memory CD4+ T cells (Luminex assay).
  • FIG. 8D shows analysis of cytokine secretion of CD19-CAR T-cell lines from sort purified na ⁇ ve, central and effector memory CD4+ T cells (Luminex assay).
  • the cytokine profile obtained by multiplex cytokine analysis ( FIG. 8B ) and proliferative capacity by CFSE staining ( FIG.
  • CD4+ ROR1-CAR modified T cells derived from the na ⁇ ve subset produced the highest levels of Th1 cytokines and proliferated most vigorously after stimulation with ROR1-positive tumor cells, suggesting they may be best suited to augment CD8+ ROR1-CAR CTLs.
  • Analysis of cytokine secretion of CD19-CAR T cell lines from sort-purified na ⁇ ve, central and effector memory CD4+ T cells demonstrates that the activity of CD4 T cell subsets is generalizable to many CARs.
  • FIG. 9 shows co-culture of CD8+ ROR1-CAR modified T cells with CD4+ ROR1-CAR modified T cells (but not untransduced control CD4+ T cells).
  • CD4 na ⁇ ve ROR1-CAR T cells provide the greatest proliferation of CD8 central memory ROR1-CAR CTLs.
  • Co culture leads to an increase in tumor-specific proliferation of the CD8+ subset, and that maximum proliferation of the CD8+ subset is observed after co-culture with CD4+ ROR1-CAR T cells derived from na ⁇ ve CD4+ T cells.
  • FIG. 10 shows the superior ability of CD4+ CAR T-cell lines derived from the na ⁇ ve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1.
  • CD4+ CAR T-cell lines derived from the na ⁇ ve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL was confirmed in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1.
  • FIGS. 11A and 11B show that CD8+ CAR T cells and CD4+ CAR T cells independently confer direct anti-tumor efficacy in a lymphoma model in immunodeficient mice (NOD/SCID-Raji).
  • Mice received either CD19-CAR transduced or control mock-transduced CD8+ central memory-derived ( FIG. 11A ), or CD19-CAR transduced or control mock-transduced CD4+ na ⁇ ve-derived T cells ( FIG. 11B ). Tumor burden and distribution was analyzed using serial bioluminescence imaging.
  • FIG. 12 shows the augmentation and synergistic effect CD4+ ROR1-CAR modified T cells on the anti-tumor efficacy of CD8+ ROR1-CAR CTLs in a mouse tumor model of systemic mantle cell lymphoma (NSG/Jeko-1-ffLuc).
  • FIGS. 13A-13D show synergy of CD8+ and CD4+ CD19-CAR T cells in a mouse model of systemic lymphoma (NSG/Raji).
  • NSG mice were inoculated with firefly-luciferase transduced Raji tumor cells. Engraftment of the Raji tumor was confirmed by bioluminescence imaging on day 6 after tumor inoculation (before treatment) (treatment scheme shown in FIG. 13A , tumor engraftment by bioluminescence shown in FIG. 13B .
  • mice treated with a combined CD8+ and CD4+ CAR T-cell product significantly higher levels CD8+ CAR T cells after the tumor challenge ( FIG.
  • T cells or “T lymphocytes” as used herein may be from any mammalian, preferably primate, species, including monkeys, dogs, and humans.
  • the T cells are allogeneic (from the same species but different donor) as the recipient subject; in some embodiments the T cells are autologous (the donor and the recipient are the same); in some embodiments the T cells arc syngeneic (the donor and the recipients are different but are identical twins).
  • Cytotoxic T lymphocyte refers to a T lymphocyte that expresses CD8 on the surface thereof (i.e., a CD8 + T cell).
  • such cells are preferably “memory” T cells (T M cells) that are antigen-experienced.
  • Central memory T cell refers to an antigen experienced CTL that expresses CD62L and CD45RO on the surface thereof, and does not express or has decreased expression of CD45RA as compared to na ⁇ ve cells.
  • central memory cells are positive for expression CD62L, CCR7, CD28, CD127, CD45RO, and CD95, and have decreased expression of CD54RA as compared to na ⁇ ve cells.
  • Effective memory T cell refers to an antigen experienced CTL that does not express or has decreased expression of CD62L on the surface thereof as compared to central memory cells, and does not express or has decreased expression of CD45RA as compared to na ⁇ ve cell.
  • effector memory cells are negative for expression CD62L, CCR7, CD28, CD45RA, and are positive for CD127 as compared to na ⁇ ve cells or central memory cells.
  • na ⁇ ve T cells refers to a non-antigen experienced T lymphocyte that expresses CD62L and CD45RA, and does not express or has decreased expression of CD45RO ⁇ as compared to central memory cells.
  • na ⁇ ve CD8+ T lymphocytes are characterized by the expression of phenotypic markers of na ⁇ ve T cells including CD62L, CCR7, CD28, CD3, CD127, and CD45RA.
  • T E T cells refers to a antigen experienced cytotoxic T lymphocyte cells that do not express or have decreased expression of CD62L, CCR7, CD28, and are positive for granzyme B and perforin as compared to central memory cells.
  • “Enriched” and “depleted” as used herein to describe amounts of cell types in a mixture refers to the subjecting of the mixture of the cells to a process or step which results in an increase in the number of the “enriched” type and a decrease in the number of the “depleted” cells.
  • a mixture or composition may contain 60, 70, 80, 90, 95, or 99 percent or more (in number or count) of the “enriched” cells and 40, 30, 20, 10, 5 or 1 percent or less (in number or count) of the “depleted” cells.
  • Interleukin-15 is a known and described in, for example, U.S. Pat. No. 6,344,192.
  • CAR refers to chimeric antigen receptor comprising an extracellular variable domain of an antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as a costimulatory domain.
  • CD4+ T lymphocytes during in vitro culture significantly increase proliferation, persistence and anti-tumor reactivity of tumor-specific CD8+ T cells in vitro and in vivo.
  • na ⁇ ve CD4+ T cells possess an intrinsic programming that leads to superior helper activity compared to CD4+ T cells derived from central and effector memory, or bulk CD4+ T cells.
  • tumor-reactive CD4+ T cells are modified with a single-chain antibody-derived chimeric antigen receptor (CAR) specific for the orphan tyrosine kinase receptor ROR1 or for the CD19 molecule.
  • CAR single-chain antibody-derived chimeric antigen receptor
  • ROR1 is uniformly expressed on chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) and ROR1-specific CAR from an anti-ROR1 monoclonal antibody (mAb) confers specific recognition of malignant, but not mature normal B-cells when expressed in CD8+ cytotoxic T cells (CTLs).
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • mAb anti-ROR1 monoclonal antibody
  • ROR1-CAR T cells from bulk and flow sort purified na ⁇ ve, central and effector memory CD4+ T cells are obtained from the peripheral blood of both healthy donors and CLL patients.
  • CD4+ CAR T cells had specific but weak cytolytic activity against ROR1+ tumors including primary CLL, the MCL line Jeko-1, and K562 cells transfected with ROR1.
  • Multiplex cytokine analysis detects high-level production of Th1 cytokines with significantly higher levels of IFN ⁇ , TNF ⁇ , and particularly IL-2 compared to CD8+ CAR CTLs.
  • CFSE staining shows dramatically higher proliferation after stimulation with ROR1-positive tumor cells, with both the percentage of cells that were induced to proliferate and the number of cell divisions that the proliferating subset underwent being significantly higher compared to CD8+ CAR CTL.
  • CD4+ T cells obtained from both healthy donors and CLL patients acquire anti-tumor reactivity after genetic modification with a ROR1-specific CAR.
  • the ability to proliferate in the absence of exogenous cytokines and to produce high levels of Th1 cytokines demonstrates that CD4+ CAR T cells exert typical helper functions after stimulation through the CAR and suggests that in addition to conferring direct anti-tumor effects, could be utilized to augment tumor-specific CD8+ CTL.
  • the cytokine profile and proliferative capacity of ROR1-CAR T cells derived from flow sort purified CD4+ na ⁇ ve, central and effector memory subsets is obtained.
  • the CD4+ CAR T cells derived from the na ⁇ ve CD45RA+ CD45RO ⁇ CD62L+ subset, produces the highest levels of Th1 cytokines, especially IL-2, and proliferates in response to ROR1+ tumor cells. Indeed, in co-culture experiments, the addition of CAR-transduced, but not untransduced CD4+ T cells leads to a significant increase in tumor-specific proliferation of CD8+ CAR CTLs.
  • CAR-modified CD4+T cells derived from na ⁇ ve rather than central and effector memory subsets or bulk CD4+ T cells results in enhanced proliferation of CD8+ CAR CTL.
  • CD8+ central memory T cells have an intrinsic programming that allows them to persist for extended periods after administration, which makes them the preferred subset of CD8+ T cells for immunotherapy.
  • ROR1-CAR or CD19 CAR modified CTLs from sort purified CD8+ central memory T cells and CD4+ na ⁇ ve CAR-modified T cells provide enhanced proliferation of the CD8+ T cell subset.
  • tumor-specific CD4+ T cells exert anti-tumor reactivity and provide help to tumor-specific CD8+ T cells in vitro and in vivo.
  • tumor-specific CD4+ T cells from the na ⁇ ve subset are utilized.
  • the CD8+ and CD4+ T cells can be modified with a T cell receptor (TCR).
  • TCR T cell receptor
  • the TCR could be specific for any antigen, pathogen or tumor (there are TCRs for many tumor antigens in melanoma (MART1, gp100 for example), leukemia (WT1, minor histocompatibility antigens for example), breast cancer (her2, NY-BR1 for example).
  • the disclosure provides for an adoptive cellular immunotherapy composition
  • a genetically modified helper T lymphocyte cell preparation that augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response
  • the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor or other receptors.
  • an adoptive cellular immunotherapy composition further comprises a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • an adoptive cellular immunotherapy composition comprises a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, in combination with an antigen-reactive chimeric antigen receptor modified na ⁇ ve CD4+ T helper cell derived from CD45RO negative, CD62L positive CD4 positive T cells, and a pharmaceutically acceptable carrier.
  • an adoptive cellular immunotherapy composition comprises an antigen specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response derived from the patient combined with an antigen-reactive chimeric antigen receptor modified na ⁇ ve CD4+ T helper cell that augments the CD8+ immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • an adoptive cellular immunotherapy composition comprises an antigen-reactive chimeric antigen receptor modified na ⁇ ve CD4+ T helper cell that augments the CD8+ immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with a disease or disorder and an intracellular signaling domain of a T cell receptor.
  • the CD4+ T helper lymphocyte cell is selected from the group consisting of na ⁇ ve CD4+ T cells, central memory CD4+ T cells, effector memory CD4+ T cells, or bulk CD4+ T cells.
  • CD4+ helper lymphocyte cell is a na ⁇ ve CD4+ T cell, wherein the na ⁇ ve CD4+ T cell comprises a CD45RO ⁇ , CD45RA+, CD62L+ CD4+ T cell.
  • the CD8+ T cytotoxic lymphocyte cell is selected from the group consisting of na ⁇ ve CD8+ T cells, central memory CD8+ T cells, effector memory CD8+ T cells or bulk CD8+ T cells.
  • the CD8+ cytotoxic T lymphocyte cell is a central memory T cell wherein the central memory T cell comprises a CD45RO+, CD62L+, CD8+ T cell.
  • the CD8+ cytotoxic T lymphocyte cell is a central memory T cell and the CD4+ helper T lymphocyte cell is a na ⁇ ve CD4+ T cell.
  • the T cells can be modified with a recombinant T cell receptor.
  • TCR could be specific for any antigen, pathogen or tumor.
  • TCRs for many tumor antigens in melanoma MART1, gp100, for example
  • WT1 leukemia
  • minor histocompatibility antigens for example
  • breast cancer her2, NY-BR1, for example.
  • compositions described herein provide for antigen reactive CD4+ and CD8+ T lymphocytes.
  • T lymphocytes can be collected in accordance with known techniques and enriched or depleted by known techniques such as affinity binding to antibodies such as flow cytometry and/or immunomagnetic selection. After enrichment and/or depletion steps, in vitro expansion of the desired T lymphocytes can be carried out in accordance with known techniques (including but not limited to those described in U.S. Pat. No. 6,040,177 to Riddell et al.), or variations thereof that will be apparent to those skilled in the art.
  • the desired T cell population or subpopulation may be expanded by adding an initial T lymphocyte population to a culture medium in vitro, and then adding to the culture medium feeder cells, such as non-dividing peripheral blood mononuclear cells (PBMC) (e.g., such that the resulting population of cells contains at least about 5, 10, 20, or 40 or more PBMC feeder cells for each T lymphocyte in the initial population to be expanded); and incubating the culture (e.g., for a time sufficient to expand the numbers of T cells).
  • the non-dividing feeder cells can comprise gamma-irradiated PBMC feeder cells.
  • the PBMC are irradiated with gamma rays in the range of about 3000 to 3600 rads.
  • the order of addition of the T cells and feeder cells to the culture media can be reversed if desired.
  • the culture can typically be incubated under conditions of temperature and the like that are suitable for the growth of T lymphocytes.
  • the temperature will generally be at least about 25 degrees Celsius, preferably at least about 30 degrees, more preferably about 37 degrees.
  • the T lymphocytes expanded include cytotoxic T lymphocytes (CTL) and helper T lymphocytes that are specific for an antigen present on a human tumor or a pathogen.
  • CTL cytotoxic T lymphocytes
  • helper T lymphocytes that are specific for an antigen present on a human tumor or a pathogen.
  • the expansion method may further comprise the step of adding non-dividing EBV-transformed lymphoblastoid cells (LCL) as feeder cells.
  • LCL can be irradiated with gamma rays in the range of about 6000 to 10,000 rads.
  • the LCL feeder cells may be provided in any suitable amount, such as a ratio of LCL feeder cells to initial T lymphocytes of at least about 10:1.
  • the expansion method may further comprise the step of adding anti-CD3 monoclonal antibody to the culture medium (e.g., at a concentration of at least about 0.5 ng/ml).
  • the expansion method may further comprise the step of adding IL-2 and/or IL-15 to the culture medium (e.g., wherein the concentration of IL-2 is at least about 10 units/ml).
  • both cytotoxic and helper T lymphocytes can be sorted into na ⁇ ve, memory, and effector T cell subpopulations either before or after expansion.
  • CD8+ cells can be obtained by using standard methods.
  • CD8+ cells are further sorted into na ⁇ ve, central memory, and effector cells by identifying cell surface antigens that are associated with each of those types of CD8+ cells.
  • memory T cells are present in both CD62L+ and CD62L ⁇ subsets of CD8+ peripheral blood lymphocytes.
  • PBMC are sorted into CD62L-CD8+ and CD62L+ CD8+ fractions after staining with anti-CD8 and anti-CD62L antibodies.
  • the expression of phenotypic markers of central memory T CM include CD45RO, CD62L, CCR7, CD28, CD3, and CD127 and are negative for granzyme B.
  • central memory T cells are CD45RO+, CD62L+, CD8+ T cells.
  • effector T E are negative for CD62L, CCR7, CD28, and CD127, and positive for granzyme B and perforin.
  • na ⁇ ve CD8+ T lymphocytes are characterized by the expression of phenotypic markers of na ⁇ ve T cells including CD62L, CCR7, CD28, CD3, CD127, and CD45RA.
  • Whether a cell or cell population is positive for a particular cell surface marker can be determined by flow cytometry using staining with a specific antibody for the surface marker and an isotype matched control antibody.
  • a cell population negative for a marker refers to the absence of significant staining of the cell population with the specific antibody above the isotype control, positive refers to uniform staining of the cell population above the isotype control.
  • a decrease in expression of one or markers refers to loss of 1 log 10 in the mean fluorescence intensity and/or decrease of percentage of cells that exhibit the marker of at least 20% of the cells, 25% of-the cells, 30% of the cells, 35% of the cells, 40% of the cells, 45% of the cells, 50% of the cells, 55% of the cells, 60% of the cells, 65% of the cells, 70% of the cells, 75% of the cells, 80% of the cells, 85% of the cells, 90% of the cell, 95% of the cells, and 100% of the cells and any % between 20 and 100% when compared to a reference cell population.
  • a cell population positive for of one or markers refers to a percentage of cells that exhibit the marker of at least 50% of the cells, 55% of the cells, 60% of the cells, 65% of the cells, 70% of the cells, 75% of the cells, 80% of the cells, 85% of the cells, 90% of the cell, 95% of the cells, and 100% of the cells and any % between 50 and 100% when compared to a reference cell population.
  • CD4+ T helper cells are sorted into na ⁇ ve, central memory, and effector cells by identifying cell populations that have cell surface antigens.
  • CD4+ lymphocytes can be obtained by standard methods.
  • na ⁇ ve CD4+ T lymphocytes are CD45RO ⁇ , CD45RA+, CD62L+ CD4+ T cell.
  • central memory CD4+ cells are CD62L positive and CD45RO positive.
  • effector CD4+ cells are CD62L and CD45RO negative.
  • CD4+ and CD8+ that are antigen specific can be obtained by stimulating na ⁇ ve or antigen specific T lymphocytes with antigen.
  • antigen specific T cell clones can be generated to Cytomegalovirus antigens by isolating T cells from infected subjects and stimulating the cells in vitro with the same antigen. Na ⁇ ve T cells may also be used. Any number of antigens from tumor cells, cancer cells, or infectious agents may be utilized.
  • the adoptive cellular immunotherapy compositions are useful in the treatment of a disease or disorder including a solid tumor, hematologic malignancy, melanoma, or infection with a virus.
  • the introduced gene or genes may improve the efficacy of therapy by promoting the viability and/or function of transferred T cells; or they may provide a genetic marker to permit selection and/or evaluation of in vivo survival or migration; or they may incorporate functions that improve the safety of immunotherapy, for example, by making the cell susceptible to negative selection in vivo as described by Lupton S. D. et al., Mol.
  • T cells are modified with chimeric antigen receptors (CAR).
  • CARs comprise a single-chain antibody fragment (scFv) that is derived from the variable heavy (VH) and variable light (VL) chains of a monoclonal antibody (mAb) linked to the TCR CD3+ chain that mediates T-cell activation and cytotoxicity.
  • Costimulatory signals can also be provided through the CAR by fusing the costimulatory domain of CD28 or 4-1BB to the CD3+ chain.
  • CARs are specific for cell surface molecules independent from HLA, thus overcoming the limitations of TCR-recognition including HLA-restriction and low levels of HLA-expression on tumor cells.
  • CARs can be constructed with a specificity for any cell surface marker by utilizing antigen binding fragments or antibody variable domains of, for example, antibody molecules.
  • the antigen binding molecules can be linked to one or more cell signaling modules.
  • cell signaling modules include CD3 transmembrane domain, CD3 intracellular signaling domains, and CD 28 transmembrane domains.
  • the intracellular signaling domain comprises a CD28 transmembrane and signaling domain linked to a CD3 intracellular domain.
  • a CAR can also include a transduction marker, such as tEGFR.
  • the intracellular signaling domain of the CD8+ cytotoxic T cells is the same as the intracellular signaling domain of the CD4+ helper T cells. In other embodiments, the intracellular signaling domain of the CD8+ cytotoxic T cells is different than the intracellular signaling domain of the CD4+ helper T cells.
  • the CD8+ T cell and the CD4+ T cell are both genetically modified with an antibody heavy chain domain that specifically binds a pathogen-specific cell surface antigen.
  • CARs are specific for cell surface expressed antigens associated with pathogens, tumors, or cancer cells.
  • a CAR is specific for HIV antigens, HCV antigens, HBV antigens, CMV antigens, parasitic antigens, and tumor antigens such as orphan tyrosine kinase receptor ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, and CEA.
  • Methods for producing a CAR are described herein and can also be found in U.S.
  • the same or a different CAR can be introduced into each of CD4+ and CD8+ T lymphocytes.
  • the CAR in each of these populations has an antigen binding molecule that specifically binds to the same antigen.
  • the cellular signaling modules can differ.
  • each of the CD4 or CD8 T lymphocytes can be sorted in to na ⁇ ve, central memory, effector memory or effector cells prior to transduction.
  • each of the CD4 or CD8 T lymphocytes can be sorted in to na ⁇ ve, central memory, effector memory, or effector cells prior to transduction.
  • the T cells can be modified with a recombinant T cell receptor.
  • TCR could be specific for any antigen, pathogen or tumor.
  • TCRs for many tumor antigens in melanoma MART1, gp100 for example
  • WT1 leukemia
  • minor histocompatibility antigens for example breast cancer (her2, NY-BR1 for example).
  • viral vectors which have been used in this way include virus vectors derived from simian virus 40, adenoviruses, adeno-associated virus (AAV), lentiviral vectors, and retroviruses.
  • virus vectors derived from simian virus 40 include virus vectors derived from simian virus 40, adenoviruses, adeno-associated virus (AAV), lentiviral vectors, and retroviruses.
  • AAV adeno-associated virus
  • retroviruses retroviruses
  • hematopoietic or lymphoid cells including calcium phosphate transfection, protoplast fusion, electroporation, and infection with recombinant adenovirus, adeno-associated virus and retrovirus vectors.
  • Primary T lymphocytes have been successfully transduced by electroporation and by retroviral infection.
  • Retroviral vectors provide a highly efficient method for gene transfer into eukaryotic cells. Moreover, retroviral integration takes place in a controlled fashion and results in the stable integration of one or a few copies of the new genetic information per cell.
  • a stimulatory factor for example, a lymphokine or a cytokine
  • a stimulatory factor for example, a lymphokine or a cytokine
  • gene segments that cause the T cells of the invention to be susceptible to negative selection in vivo By “negative selection” is meant that the infused cell can be eliminated as a result of a change in the in vivo condition of the individual.
  • the negative selectable phenotype may result from the insertion of a gene that confers sensitivity to an administered agent, for example, a compound.
  • Negative selectable genes include, inter alia the following: the Herpes simplex virus type I thymidine kinase (HSV-I TK) gene (Wigler et al., Cell 11:223, 1977) which confers ganciclovir sensitivity; the cellular hypoxanthine phosphribosyltransferase (HPRT) gene, the cellular adenine phosphoribosyltransferase (APRT) gene, bacterial cytosine deaminase, (Mullen et al., Proc. Natl. Acad. Sci. USA. 89:33, 1992).
  • HSV-I TK Herpes simplex virus type I thymidine kinase
  • HPRT hypoxanthine phosphribosyltransferase
  • APRT cellular adenine phosphoribosyltransferase
  • the T cells may be useful to include in the T cells a positive marker that enables the selection of cells of the negative selectable phenotype in vitro.
  • the positive selectable marker may be a gene which, upon being introduced into the host cell expresses a dominant phenotype permitting positive selection of cells carrying the gene.
  • Genes of this type are known in the art, and include, inter alia, hygromycin-B phosphotransferase gene (hph) which confers resistance to hygromycin B, the amino glycoside phosphotransferase gene (neo or aph) from Tn5 which codes for resistance to the antibiotic G418, the dihydrofolate reductase (DHFR) gene, the adenosine daminase gene (ADA), and the multi-drug resistance (MDR) gene.
  • hph hygromycin-B phosphotransferase gene
  • DHFR dihydrofolate reductase
  • ADA adenosine daminase gene
  • MDR multi-drug resistance
  • the positive selectable marker and the negative selectable element are linked such that loss of the negative selectable element necessarily also is accompanied by loss of the positive selectable marker.
  • the positive and negative selectable markers are fused so that loss of one obligatorily leads to loss of the other.
  • An example of a fused polynucleotide that yields as an expression product a polypeptide that confers both the desired positive and negative selection features described above is a hygromycin phosphotransferase thymidine kinase fusion gene (HyTK). Expression of this gene yields a polypeptide that confers hygromycin B resistance for positive selection in vitro, and ganciclovir sensitivity for negative selection in vivo.
  • the polynucleotides of the invention encoding the chimeric receptors are in retroviral vectors containing the fused gene, particularly those that confer hygromycin B resistance for positive selection in vitro, and ganciclovir sensitivity for negative selection in vivo, for example the HyTK retroviral vector described in Lupton, S. D. et al. (1991), supra. See also the publications of PCT/US91/08442 and PCT/US94/05601, by S. D. Lupton, describing the use of bifunctional selectable fusion genes derived from fusing a dominant positive selectable marker with negative selectable markers.
  • Preferred positive selectable markers are derived from genes selected from the group consisting of hph, nco, and gpt
  • preferred negative selectable markers are derived from genes selected from the group consisting of cytosine deaminase, HSV-I TK, VZV TK, HPRT, APRT and gpt.
  • Especially preferred markers are bifunctional selectable fusion genes wherein the positive selectable marker is derived from hph or neo, and the negative selectable marker is derived from cytosine deaminase or a TK gene or selectable marker.
  • retroviral transductions can be carried out as follows: on day 1 after stimulation using REM as described herein, provide the cells with 20-30 units/ml IL-2; on day 3, replace one half of the medium with retroviral supernatant prepared according to standard methods and then supplement the cultures with 5 ⁇ g/ml polybrene and 20-30 units/ml IL-2; on day 4, wash the cells and place them in fresh culture medium supplemented with 20-30 units/ml IL-2; on day 5, repeat the exposure to retrovirus; on day 6, place the cells in selective medium (containing, e.g., an antibiotic corresponding to an antibiotic resistance gene provided in the retroviral vector) supplemented with 30 units/ml IL-2; on day 13, separate viable cells from dead cells using Ficoll Hypaque density gradient separation and then subclone the viable cells.
  • selective medium containing, e.g., an antibiotic corresponding to an antibiotic resistance gene provided in the retroviral vector
  • CD4+ and CD8+ cells can be modified with an expression vector encoding a CAR. In embodiments, these cells are then further sorted into subpopulations of na ⁇ ve, central memory and effector cells as described above by sorting for cell surface antigens unique to each of those cell populations.
  • CD4+ or CD8+ cell populations may be selected by their cytokine profile or proliferative activities. For example, CD4+ T lymphocytes that have enhanced production of cytokines such as IL-2, IL-4, IL-10, TNF ⁇ , and IFN ⁇ as compared to sham transduced cells or transduced CD8+ cells when stimulated with antigen can be selected. In other embodiments, na ⁇ ve CD4+ T cells that have enhanced production of IL-2 and/or TNF ⁇ are selected. Likewise, CD8+ cells that have enhanced IFN ⁇ production are selected as compared to sham transduced CD8+ cells.
  • CD4+ and CD8+ cells that proliferate in response to antigen are selected.
  • CD4+ cells that proliferate vigorously when stimulated with antigen as compared to sham transduced cells, or CD8+ transduced cells are selected.
  • CD4+ and CD8+ cells are selected that are cytotoxic for antigen bearing cells.
  • CD4+ are expected to be weakly cytotoxic as compared to CD8+ cells.
  • combinations of CD4+ and CD8+ T cells will be utilized in the compositions.
  • combinations of CAR transduced CD4+ cells can be combined with CD8+ antigen reactive cells to the same antigenic specificity as the CAR.
  • CAR transduced CD8+ cells are combined with antigen reactive CD4+ cells.
  • CAR modified CD4+ and CD8+ cells are combined.
  • CD4+ and CD8+ cells can be further separated into subpopulations, such as na ⁇ ve, central memory, and effector cell populations.
  • na ⁇ ve CD4+ cells are CD45RO ⁇ , CD45RA+, CD62L+ CD4+ T cells.
  • central memory CD4+ cells are CD62L positive and CD45RO positive.
  • effector CD4+ cells are CD62L negative and CD45RO positive. Each of these populations may be independently modified with a CAR.
  • memory T cells are present in both CD62L+ and CD62L ⁇ subsets of CD8+ peripheral blood lymphocytes.
  • PBMCs are sorted into CD62L-CD8+ and CD62L+ CD8+ fractions after staining with anti-CD8 and anti-CD62L antibodies.
  • expression of phenotypic markers of central memory T CM include CD62L, CCR7, CD28, CD3, and CD127 and are negative for granzyme B.
  • central memory T cells are CD45RO+, CD62L+, CD8+ T cells.
  • effector T E cells are negative for CD62L, CCR7, CD28, and CD127, and positive for granzyme B and perforin.
  • na ⁇ ve CD8+T lymphocytes are characterized by CD8+, CD62L+, CD45RO+, CCR7+, CD28+CD127+, and CD45RO+. Each of these populations may be independently modified with a CAR.
  • modified na ⁇ ve CD4+ cells are combined with modified central memory CD8+ T cells to provide a synergistic cytotoxic effect on antigen bearing cells, such as tumor cells.
  • the disclosure provides methods of making adoptive immunotherapy compositions and uses or methods of using these compositions for performing cellular immunotherapy in a subject having a disease or disorder.
  • a method of manufacturing the compositions comprises obtaining a modified na ⁇ ve CD4+ T helper cell, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain.
  • a method further comprises obtaining a modified CD8+ cytotoxic T cell, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • a method comprises obtaining a modified CD8+ cytotoxic T cell, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and further comprising combining the modified CD8+ cytotoxic T cells with an antigen specific CD4+ helper cell lymphocyte cell preparation.
  • Antigen specific T lymphocytes can be obtained from a patient having the disease or disorder or can be prepared by in vitro stimulation of T lymphocytes in the presence of antigen. Subpopulations of CD4+ and CD8+ T lymphocytes can also be isolated as described herein and combined in the methods of manufacturing.
  • a method comprises administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation that provides a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors and a genetically modified helper T lymphocyte cell preparation that elicits direct tumor recognition and augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
  • a method of performing cellular immunotherapy in subject having a disease or disorder comprises: administering to the subject a genetically modified helper T lymphocyte cell preparation, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising a extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor.
  • the method further comprises administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8 positive cells that have a chimeric antigen receptor comprising a extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor.
  • Another embodiment describes a method of performing cellular immunotherapy in a subject having a disease or disorder comprising: analyzing a biological sample of the subject for the presence of an antigen associated with the disease or disorder and administering the adoptive immunotherapy compositions described herein, wherein the chimeric antigen receptor specifically binds to the antigen.
  • a CAR is produced that has a component that provides for specific binding to an antigen associated with a disease or conditions, such as a solid tumor, cancer, viral infection, and an infection with a parasite.
  • the intracellular signaling module of a T cell receptor of the chimeric antigen receptor comprises a transmembrane domain, a CD28 signaling domain, and a CD3 intracellular signaling domain, or other domains of T cell costimulatory molecules.
  • the intracellular signaling molecule comprises the CD3 intracellular domain, a CD28 domain, a CD28 transmembrane and signaling domain linked to a CD3 intracellular domain, or other domains of T cell costimulatory molecules.
  • the T cells can be modified with a recombinant T cell receptor.
  • TCR could be specific for any antigen, pathogen or tumor.
  • TCRs for many tumor antigens in melanoma MART1, gp100 for example
  • WT1 leukemia
  • minor histocompatibility antigens for example breast cancer (her2, NY-BR1 for example).
  • the CD4+ T helper lymphocyte cell is selected from the group consisting of na ⁇ ve CD4+ T cells, central memory CD4+ T cells, effector memory CD4+ T cells or bulk CD4+ T cells.
  • CD4+ helper lymphocyte cell is a na ⁇ ve CD4+ T cell, wherein the na ⁇ ve CD4+ T cell comprises a CD45RO ⁇ , CD45RA+, CD62L+ CD4+ T cell.
  • the CD8+ T cytotoxic lymphocyte cell is selected from the group consisting of na ⁇ ve CD8+ T cells, central memory CD8+ T cells, effector memory CD8+ T cells or bulk CD8+ T cells.
  • the CD8+ cytotoxic T lymphocyte cell is a central memory T cell wherein the central memory T cell comprises a CD45RO+, CD62L+, CD8+ T cell.
  • the CD8+ cytotoxic T lymphocyte cell is a central memory T cell and the CD4+ helper T lymphocyte cell is a na ⁇ ve CD4+ T cell.
  • the CD8+ T cell and the CD4+ T cell are both genetically modified with a CAR comprising an antibody heavy chain domain that specifically binds a pathogen or tumor-specific cell surface antigen.
  • the intracellular signaling domain of the CD8 cytotoxic T cells is the same as the intracellular signaling domain of the CD4 helper T cells.
  • the intracellular signaling domain of the CD8 cytotoxic T cells is different than the intracellular signaling domain of the CD4 helper T cells.
  • Subjects that can be treated by the present invention are, in general, human and other primate subjects, such as monkeys and apes for veterinary medicine purposes.
  • the subjects can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects.
  • the methods are useful in the treatment of, for example, solid tumor, hematologic malignancy, melanoma, or infection with a virus or other pathogen.
  • Infections with pathogens include HIV, HCV, HBV, CMV, and parasitic disease.
  • the antigen associated with the disease or disorder is selected from the group consisting of orphan tyrosine kinase receptor ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, CEA, and hepatitis B surface antigen.
  • Subjects that can be treated include subjects afflicted with cancer, including but not limited to colon, lung, liver, breast, prostate, ovarian, skin (including melanoma), bone, and brain cancer, etc.
  • the tumor associated antigens are known, such as melanoma, breast cancer, squamous cell carcinoma, colon cancer, leukemia, myeloma, prostate cancer, etc.
  • memory T cells can be isolated or engineered by introducing the T cell receptor genes).
  • the tumor associated proteins can be targeted with genetically modified T cells expressing an engineered immunoreceptor. Examples include but are not limited to B cell lymphoma, breast cancer, prostate cancer, and leukemia.
  • Subjects that can be treated also include subjects afflicted with, or at risk of developing, an infectious disease, including but not limited to viral, retroviral, bacterial, and protozoal infections, etc.
  • Subjects that can be treated include immunodeficient patients afflicted with a viral infection, including but not limited to Cytomegalovirus (CMV), Epstein-Barr virus (EBV), adenovirus, BK polyomavirus infections in transplant patients, etc.
  • Cells prepared as described above can be utilized in methods and compositions for adoptive immunotherapy in accordance with known techniques, or variations thereof that will be apparent to those skilled in the art based on the instant disclosure. See, e.g., US Patent Application Publication No. 2003/0170238 to Gruenberg et al; see also U.S. Pat. No. 4,690,915 to Rosenberg.
  • the cells are formulated by first harvesting them from their culture medium, and then washing and concentrating the cells in a medium and container system suitable for administration (a “pharmaceutically acceptable” carrier) in a treatment-effective amount.
  • a medium and container system suitable for administration a “pharmaceutically acceptable” carrier
  • Suitable infusion medium can be any isotonic medium formulation, typically normal saline, Normosol R (Abbott) or Plasma-Lyte A (Baxter), but also 5% dextrose in water or Ringer's lactate can be utilized.
  • the infusion medium can be supplemented with human serum albumin.
  • a treatment-effective amount of cells in the composition is at least 2 cells (for example, 1 CD8+ central memory T cell and 1 CD4+ helper T cell subset) or is more typically greater than 10 2 cells, and up to 10 6 , up to and including 10 8 or 10 9 cells and can be more than 10 10 cells.
  • the number of cells will depend upon the ultimate use for which the composition is intended as will the type of cells included therein. For example, if cells that are specific for a particular antigen are desired, then the population will contain greater than 70%, generally greater than 80%, 85% and 90-95% of such cells.
  • the cells are generally in a volume of a liter or less, can be 500 mls or less, even 250 mls or 100 mls or less.
  • the density of the desired cells is typically greater than 10 6 cells/ml and generally is greater than 10 7 cells/ml, generally 10 8 cells/ml or greater.
  • the clinically relevant number of immune cells can be apportioned into multiple infusions that cumulatively equal or exceed 10 9 , 10 10 or 10 11 cells.
  • the lymphocytes of the invention may be used to confer immunity to individuals.
  • immuno is meant a lessening of one or more physical symptoms associated with a response to infection by a pathogen, or to a tumor, to which the lymphocyte response is directed.
  • the amount of cells administered is usually in the range present in normal individuals with immunity to the pathogen.
  • the cells are usually administered by infusion, with each infusion in a range of from 2 cells, up to at least 10 6 to 10 10 cells/m 2 , preferably in the range of at least 10 7 to 10 9 cells/m 2 .
  • the clones may be administered by a single infusion, or by multiple infusions over a range of time.
  • T lymphocytes including cytotoxic T lymphocytes and/or helper T lymphocytes
  • the rapid expansion method of the present invention See, e.g., U.S. Pat. No. 6,040,177 to Riddell et al. at column 17.
  • a ROR1-specific CAR can be expressed in human CD8+ T cells and confers specific recognition of ROR1+ B-cell tumors and not mature normal B cells.
  • Epstein-Barr virus transformed B cells were generated as described (25).
  • the tumor cell lines Jeko-1, and, BALL-1, were provided by Drs Oliver Press and Jerald Radich (Fred Hutchinson Cancer Research Center). All cell lines were maintained in RPMI, 10% fetal calf serum, 0.8 mM L-glutamine, and 1% penicillin-streptomycin (LCL medium).
  • K562 cells were obtained from the American Type Culture Collection.
  • Effectene transfection reagent (QIAGEN) was used to transfect Platinum-A cells (Cell Biolabs) with MIGR-1/ROR1 and produce ROR1-encoding retrovirus.
  • K562 cells were retrovirally transduced by centrifugation at 2500 rpm for 60 minutes at 32° C., expanded, and the ROR1-positive subset was sort-purified.
  • First-strand cDNA of B-CLL, normal resting and activated B cells, and EBV-LCL was prepared as described in the previous paragraph.
  • First-strand cDNA from normal tissues Human Tissue panels I/II, Blood Fractions
  • Expression of ROR1 mRNA was analyzed in duplicate and normalized to GAPDH. Amplifications were performed on an ABI Prism 7900 (Applied Biosystems) in a 50 reaction consisting of 25 ⁇ L Power SYBR Green PCR Master Mix (Applied Biosystems), 2.5 ng of cDNA, and 300 nM gene-specific forward and reverse primers:
  • ROR1-F 5-AGCGTGCGATTCAAAGGATT-3 ROR1-R 5-GACTGGTGCCGACGATGACT-3, GAPDH-F 5-GAAGGTGAAGGTCGGAGTC-3, and GAPDH-R 5-GAAGATGGTGATGGGATTTC-3.
  • the cycle threshold (Ct) was determined using SDS software v2.2.2 (Applied Biosystems) and the level of gene expression calculated using the comparative Ct method (2-( ⁇ Ct)).
  • CD20-CAR CD20R-epHIV7
  • GFP-encoding lentiviral vectors GFP-epHIV7
  • the ROR1-CAR was encoded in the same vector.
  • a mouse mAb (clone 2A2) that demonstrated specific binding to human ROR1 expressed on primary B-CLL and MCL tumor lines was generated, cloned, and characterized in a previous study.
  • a codon-optimized nucleotide sequence encoding a scFv containing the VL and VH chain of mAb 2A2 was synthesized (GENEART) and cloned into CD20R-epHIV7 using NheI and RsrII restriction sites to replace the CD20-specific scFv.
  • Lentivirus was produced in 293T cells co-transfected with the lentiviral vector and the packaging vectors pCHGP-2, pCMVRev2, and pCMV-G using Effectene (Qiagen). Medium was changed 16 hours after transfection and lentivirus collected after 48 hours.
  • T CM central memory T cells
  • each transduced T-cell line was stained with biotin-conjugated anti-EGFR (epithelial growth factor receptor) mAb, streptavidin-PE, and anti-CD8 mAb.
  • EGFR+CD8+ T cells were sort purified and cloned by limiting dilution (0.5 cells/well) (25).
  • ROR1-CAR transduced T cells were identified by staining with biotinylated recombinant Fc-ROR1 extracellular domain fusion protein and streptavidin-PE.
  • Recombinant ROR1-protein was produced in transiently transfected 293F cells (Invitrogen), purified as described (26), and biotinylated using the BiotinTag kit (Sigma). GFP-transduced CD8+ T cells were identified by flow cytometry, sort-purified, and cloned in similar fashion.
  • Target cells were labeled with 51 Cr (PerkinElmer) overnight, washed and incubated in triplicate at 1-2 ⁇ 10 3 cells/well with effector T cells at various effector to target (E:T) ratios. Supernatants were harvested for ⁇ counting after a 4-hour incubation, and specific lysis was calculated using the standard formula (25).
  • Transduced CD8+ T cells were sort-purified using a biotinylated anti-EGFR mAb and streptavidin conjugated dyes.
  • ROR1-CAR expression on the surface of the sort-purified T cells was evaluated by staining the cells with a biotinylated recombinant Fc-ROR1 extracellular domain fusion protein that directly binds to the scFv of the ROR1-CAR, and costaining with streptavidin-conjugates.
  • Fc-ROR1-protein specifically stained CD8+ T cells transduced with the ROR1-CAR lentiviral vector but not CD8+ T cells transduced with a control lentiviral vector encoding GFP ( FIG. 1 ).
  • the ROR1-CAR transduced T-cell clones efficiently lysed primary B-CLL and K562 cells that were stably transfected with the ROR1-gene, but not native, ROR1-negative K562 cells, demonstrating specific recognition of ROR1 ( FIG. 2 ).
  • Adoptive immunotherapies that employ CAR-modified T cells are being investigated in clinical trials for B-cell malignancies.
  • the surface molecules that are being targeted are B-cell lineage-specific and include CD19, which is expressed on normal B-lineage cells from the pro-B-cell stage to plasma cells, and CD20, which is expressed on normal B cells from the pre-B-cell stage to memory B cells.
  • CD19 which is expressed on normal B-lineage cells from the pro-B-cell stage to plasma cells
  • CD20 which is expressed on normal B cells from the pre-B-cell stage to memory B cells.
  • an anticipated outcome of effective therapy targeting these molecules is depletion of normal B cells and B-cell precursors.
  • Gene expression profiling studies have identified genes that are preferentially or exclusively expressed by malignant but not by normal B cells and ROR1 emerged as a CLL signature gene in 2 independent analyses (27,28).
  • Specific antibodies to ROR1 developed in CLL patients after vaccination with autologous tumor cells that had been modified to express CD154 and treatment with len
  • ROR1-positive malignant cells with engineered T cells expressing a ROR1-CAR.
  • CD8+ ROR1-CAR T cells could be derived from both normal donors and CLL patients after lentiviral transduction of either bulk PBMCs or sort-purified TCM, that in animal models persist for extended periods after adoptive transfer (31).
  • ROR1-CAR transduced T cells efficiently lysed primary B-CLL, but not normal resting or activated B-cells. These T cells produced effector cytokines including TNF- ⁇ , IFN ⁇ , and IL-2, and were capable of proliferating in response to ROR1-expressing tumor cells.
  • CD4+ ROR1-CAR T cells can be generated from PBMC of healthy donors/CLL-patients.
  • a ROR1-specific CAR can be expressed in human CD4+ T cells and confers specific recognition of ROR1+ B-cell tumors but not mature normal B cells.
  • Epstein-Barr virus transformed B cells were generated as described (25).
  • the tumor cell lines Jeko-1, and BALL-1 were provided by Drs Oliver Press and Jerald Radich (Fred Hutchinson Cancer Research Center). All cell lines were maintained in RPMI, 10% fetal calf serum, 0.8 mM L-glutamine, and 1% penicillin-streptomycin (LCL medium).
  • K562 and 293T cells were obtained from the American Type Culture Collection and cultured as directed.
  • Effectene transfection reagent (QIAGEN) was used to transfect Platinum-A cells (Cell Biolabs) with MIGR-1/ROR1 and produce ROR1-encoding retrovirus.
  • K562 cells were retrovirally transduced by centrifugation at 2500 rpm for 60 minutes at 32° C., expanded, and the ROR1-positive subset was sort-purified.
  • CD20-CAR CD20R-epHIV7
  • GFP-encoding lentiviral vectors GFP-epHIV7
  • the ROR1-CAR was encoded in the same vector.
  • a mouse mAb (clone 2A2) that demonstrated specific binding to human ROR1 expressed on primary B-CLL and MCL tumor lines was generated, cloned, and characterized in a previous study.
  • a codon-optimized nucleotide sequence encoding a scFv containing the VL and VH chain of mAb 2A2 was synthesized (GENEART) and cloned into CD20R-epHIV7 using NheI and RsrII restriction sites to replace the CD20-specific scFv.
  • Lentivirus was produced in 293T cells cotransfected with the lentiviral vector and the packaging vectors pCHGP-2, pCMVRev2, and pCMV-G using Effectene (Qiagen). Medium was changed 16 hours after transfection and lentivirus collected after 48 hours.
  • CD4+ T cells were isolated from PBMC of healthy donors and activated with anti-CD3 mAb (30 ng/mL) (25), and transduced in lentiviral supernatant supplemented with 1 ⁇ g/mL polybrene (Sigma-Aldrich) and 50 IU/mL recombinant human interleukin-2 (IL-2) on day 2 and 3 after activation by centrifugation at 2500 rpm for 60 minutes at 32° C.
  • anti-CD3 mAb 30 ng/mL
  • IL-2 human interleukin-2
  • T cells were expanded in RPMI containing 10% human serum, 2 mM L-glutamine, and 1% penicillin streptomycin (CTL medium).(25) After expansion, an aliquot of each transduced T-cell line was stained with biotin-conjugated anti-EGFR (epithelial growth factor receptor) mAb, streptavidin-PE, and anti-CD4 mAb. EGFR+ CD4+ T cells were sort purified and expanded. ROR1-CAR transduced T cells were identified by staining with biotinylated recombinant Fc-ROR1 extracellular domain fusion protein and streptavidin-PE.
  • CTL medium penicillin streptomycin
  • Recombinant ROR1-protein was produced in transiently transfected 293 cells (Invitrogen), purified as described (26), and biotinylated using the BiotinTag kit (Sigma). GFP-transduced CD4+ T cells were identified by flow cytometry, sort-purified, and cloned in similar fashion.
  • Target cells were labeled with 51 Cr (PerkinElmer) overnight, washed and incubated in triplicate at 1-2 ⁇ 10 3 cells/well with effector T cells at various effector to target (E:T) ratios. Supernatants were harvested for ⁇ counting after a 4-hour incubation, and specific lysis was calculated using the standard formula (25).
  • target and effector cells were plated in triplicate wells at an E/T ratio of 2:1, and interferon IFN ⁇ , tumor necrosis factor (TNF- ⁇ ), and IL-2 were measured by multiplex cytokine immunoassay (Luminex) in supernatant removed after a 24-hour incubation.
  • Luminex multiplex cytokine immunoassay
  • T cells were labeled with 0.2 ⁇ M carboxyfluorescein succinimidyl ester (CFSE; Invitrogen), washed, and plated with stimulator cells at a ratio of 2:1 in CTL medium containing 10 U/mL recombinant human IL-2. After a 72-hour incubation, cells were labeled with anti-CD4 mAb and propidium iodide (PI) to exclude dead cells from analysis. Samples were analyzed by flow cytometry, and cell division of live CD4+ T cells assessed by CFSE dilution.
  • CFSE carboxyfluorescein succinimidyl ester
  • ROR1-CAR transduced CD4+ T cells and ROR1-CAR transduced CD8+ cytotoxic T lymphocytes were labeled with CFSE, and co-cultured at a 2:1, 1:1 and 1:2 ratio. The co-cultures were then stimulated with K562/ROR1 cells and control K562 cells and cell proliferation measured by CFSE dye dilution assay after 5 days of incubation. For flow analysis, samples were stained with conjugated anti-CD8 and anti-CD4 mAb to distinguish CD8+ and CD4+ subsets.
  • CD4 + ROR1-CAR T cells from PBMC of healthy donors and CLL patients
  • ROR1 an oncofetal tyrosine kinase receptor
  • CD8 + T cells CD8 + T cells
  • this vector we encoded a truncated EGFR (epithelial growth factor receptor, tEGFR) domain downstream of the ROR1-CAR and a self-cleavable 2A element, to serve both as transduction marker and for the enrichment of transgene expressing T cells with anti-EGFR mAb ( FIG. 3 ).
  • EGFR epidermal growth factor receptor
  • Multiplex cytokine analysis revealed production of other Th1 cytokines such as TNF- ⁇ and IL-2 at significantly higher levels compared to CD8 + CAR CTL, and production of IL-4, IL-10 and IL-17 ( FIG. 5B ).
  • CD4 + CAR T cells showed dramatic and specific proliferation in response to ROR1-positive tumor cells. Both the percentage of T cells that was induced to proliferate and the number of cell divisions that the proliferating subset performed was significantly higher in CD4 + compared to CD8 + CAR T cells ( FIG. 6 ).
  • CD4 + CAR T cells exert typical helper functions after stimulation through the CAR and in addition to conferring direct anti-tumor effects, could also be utilized to augment CD8 + CAR CTL.
  • CD4 + CAR T cells were able to provide help to CD8 + CAR CTL.
  • CAR-transduced and control untransduced polyclonal CD4 + and CD8 + T cell lines that we established from healthy donors and CLL patients.
  • CD8 + T cells we defined an improvement in tumor-specific CD8 + effector function in the presence of CD4 + T cells compared to CD8 + T cells cultured alone.
  • CD4:CD8 ratios 2:1, 1:1, 1:2
  • CD8 + CAR CTL significantly increased specific proliferation of the CD8 + subset compared to CD8 + CAR CTL alone ( FIG. 7 ).
  • the increase in proliferation was most pronounced, when at least an equivalent amount of CD4 + CART cells (CD4:CD8 ratio of 2:1 or 1:1) was added to the co-culture.
  • CD4:CD8 ratio of 2:1 or 1:1 was added to the co-culture.
  • the combination of untransduced CD4 + with untransduced CD8 + T cells served as additional control and did not induce unspecific proliferation in the CD8 + subset (data not shown).
  • CD8 and CD4+ ROR1-CAR T cells could be derived from normal donors after lentiviral transduction of either bulk PBMCs or sort-purified T cells.
  • CD8+ ROR1-CAR transduced T cells efficiently lysed primary B-CLL, but not normal resting or activated B-cells.
  • CD4+ ROR1-CAR transduced T cells weakly lysed primary B-CLL, but not normal resting or activated B-cells. These T cells produced effector cytokines including TNF- ⁇ , IFN ⁇ , IL-2, IL-4, and IL-10. CAR-transduced CD4+ T cells produced significantly higher amounts of cytokines than the transduced CD8+ cells. Both cell types were capable of proliferating in response to ROR1-expressing tumor cells. Again, CD4+ ROR1-CAR T cells proliferated 2-3 fold higher than CD8+ ROR1-CAR CTLs. These results indicate that the transduced CD4+ helper T cells exert typical helper functions suggesting they could be utilized to augment CD8+ CAR CTLs.
  • CD4 T cells derived from na ⁇ ve, central and effector memory subsets and then modified with the ROR1 CAR were compared.
  • CD4+ T cells were isolated from PBMC of a healthy donor using negative magnetic bead selection (Miltenyi CD4 isolation kit) that yields untouched CD4+ T cells.
  • the CD4+ fraction was labeled with conjugated anti-CD45RA, anti-CD45RO and anti-CD62L mAb and flow sort purified using a FACS Aria flow sorter (BD Biosciences), and na ⁇ ve (CD45RA+ CD45RO ⁇ CD62L+), central memory (CD45RA ⁇ CD45RO+ CD62L+) and effector memory (CD45RA ⁇ CD45RO+ CD62L ⁇ ) CD4+ T cells purified based on expression of these defined markers.
  • T cells were labeled with 0.2 ⁇ M carboxyfluorescein succinimidyl ester (CFSE; Invitrogen), washed, and plated with stimulator cells at a ratio of 2:1 in CTL medium containing 10 U/mL recombinant human IL-2. After a 72-hour incubation, cells were labeled with anti-CD8 or CD4 mAb and propidium iodide (PI) to exclude dead cells from analysis. Samples were analyzed by flow cytometry, and cell division of live CD8+ and CD4+ T cells assessed by CFSE dilution.
  • CFSE carboxyfluorescein succinimidyl ester
  • target and effector cells were plated in triplicate wells at an E/T ratio of 2:1, and interferon INF ⁇ , tumor necrosis factor (TNF- ⁇ ), and IL-2 were measured by multiplex cytokine immunoassay (Luminex) in supernatant removed after a 24-hour incubation.
  • Luminex multiplex cytokine immunoassay
  • CD4 + N, central (CM) and effector memory (EM) CD4 + T cells from the peripheral blood of 3 healthy donors based on expression of CD45RA, CD45RO and CD62L ( FIG. 8A ), and compared their effector function after modification with the ROR1-CAR.
  • Multiparameter flow cytometry after enrichment of transgene expressing T cells showed expression of CD45RO and loss of CD45RA in the CD4 + N CAR T cell line, consistent with an activated phenotype after the lentiviral transduction.
  • the CD4 + N, CM and EM CAR T cell lines retained differential expression of CD62L, confirming that the initial flow sort purification had been performed with high purity.
  • CD4 + CAR T cells derived from N, CM and EM subsets were compared them to the CAR T cell lines generated from bulk CD4+ T cells.
  • CD8 and CD4+ ROR1-CAR T cells could be derived from both normal donors after lentiviral transduction of either bulk PBMCs and sort-purified T cells from defined na ⁇ ve or memory T cell subsets.
  • CD4+ na ⁇ ve, central memory, and effector T cells produced effector cytokines including TNF ⁇ , IFN ⁇ , IL-2, IL-4, and IL-10.
  • CAR-transduced CD4+ cells derived from the na ⁇ ve subset produced significantly higher amounts of TNF ⁇ and IL-2 than central and effector memory derived CD4+ CAR T cells after signaling through the CAR.
  • CD4 cell types were capable of proliferating in response to ROR1/K562, however in the CAR-transduced CD4+ cells derived from the na ⁇ ve subset, the percentage of T cells that was induced to proliferate and the number of cell divisions that the proliferating subset underwent were significantly higher. Both cytokine profile and proliferative capacity indicate that na ⁇ ve CD4+ ROR1-CAR T cells may be best suited to augment CD8+ ROR1-CAR CTL.
  • CD4 + Na ⁇ ve CAR T Cells have a Superior Ability to Augment the Effector Function of CD8 + CAR CTL
  • CD8 + CAR CTLs from sort purified N and CM T cells to determine the optimal combination of CD8 + and CD4 + CAR T cell subsets.
  • lentiviral transduction and enrichment of CAR-transduced CD8 + T cells using the tEGFR marker we confirmed tumor-reactivity of the CD8 + N, and CM CAR CTLs (data not shown) and performed co-culture experiments with CD4 + CAR T cells as before.
  • NSG mice Sub-lethally irradiated NOD/SCID/gamma ⁇ / ⁇ mice were engrafted via tail vein injection with 5 ⁇ 10 5 Jeko-1 cells that had been stably transfected with firefly luciferase (Jeko-1/ffLuc) to enable assessment of tumor burden and distribution using bioluminescence imaging.
  • Jeko-1/ffLuc firefly luciferase
  • mice Following tumor engraftment, groups of 3 mice received either CD8 + CAR CTLs (group 1), CD4 + CART cells (group 2), a combination of CD8 + and CD4 + ROR1-CAR transduced T cells (group 3), untransduced control T cells (group 4,5,6) via tail vein injection or no treatment (group 7).
  • the total number of transferred T cells was 10 ⁇ 10 6 in all cases.
  • CD8 + /CD4 + CAR T cell combination was greater than that of the CD8 + CAR CTL and CD4 + CAR T cell groups combined suggesting that CD4 + CAR T cells and CD8 + CAR CTL were working synergistically.
  • CD19 CART cells can be prepared as described in US 2008/0131415, which is hereby incorporated by reference.
  • CD19-CAR transduced CD4+ T cells and CD19-CAR transduced CD8+ cytotoxic T lymphocytes were labeled with CFSE, and co-cultured at a 2:1, 1:1 and 1:2 ratio. The co-cultures were then stimulated with K562/ROR1 cells and control K562 cells and cell proliferation measured by CFSE dye dilution assay after 5 days of incubation. For flow analysis, samples were stained with conjugated anti-CD8 and anti-CD4 mAb to distinguish CD8+ and CD4+ subsets.
  • NSG mice Sublethally irradiated NOD/SCID/gamma ⁇ / ⁇ (NSG) mice were engrafted via tail vein injection with 5 ⁇ 10 5 Jeko-1 cells that had been stably transfected with firefly luciferase (Jeko-1/ffLuc) to enable assessment of tumor burden and distribution using bioluminescence imaging.
  • Jeko-1/ffLuc firefly luciferase
  • mice Following tumor engraftment, groups of 3 mice received either CD8 + CD19 CAR CTLs (group 1), CD4 + CD 19 CART cells (group 2), a combination of CD8 + and CD4 + CD19CAR transduced T cells (group 3), untransduced control T cells (group 4,5,6) via tail vein injection or no treatment (group 7).
  • the total number of transferred T cells was 10 ⁇ 10 6 in all cases.
  • FIG. 10 shows the superior ability of CD4+ CAR T-cell lines derived from the na ⁇ ve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1.
  • CD4+ CAR T-cell lines derived from the central or effector memory subset augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL to much less extent.
  • FIG. 11 shows that CD8+ CAR T cells and CD4+ CAR T cells independently confer direct anti-tumor efficacy in a lymphoma model in immunodeficient mice (NOD/SCID-Raji). Mice received either CD19-CAR transduced or control mock-transduced CD8+ central memory-derived ( FIG. 11A ), or CD19-CAR transduced or control mock-transduced CD4+ na ⁇ ve-derived T cells ( FIG. 11B ).
  • FIG. 12 shows the augmentation and synergistic effect CD4+ ROR1-CAR modified T cells on the anti-tumor efficacy of CD8+ ROR1-CAR CTLs in a mouse tumor model of systemic mantle cell lymphoma (NSG/Jeko-1-ffLuc).
  • Anti-tumor efficacy of ROR1-CAR modified CD8+ and CD4+ T cells in a mouse tumor model of systemic aggressive mantle cell lymphoma (NSG/Jeko-1) was enhanced as compared to either cell population alone or when compared to untransduced cells.
  • FIG. 13 shows synergy of CD8+ and CD4+ CD19-CAR T cells in a mouse model of systemic lymphoma (NSG/Raji). Engraftment of the Raji tumor was confirmed by bioluminescence imaging on day 6 after tumor inoculation (before treatment) (treatment scheme shown in FIG. 13A , tumor engraftment by bioluminescence shown in FIG. 13B ). Analysis of tumor burden using bioluminescence imaging showed complete eradication of the Raji tumors in the cohorts of mice treated with CD8+ CD19-CAR T cells, and in mice treated with the combined CD8+ and CD4+ CD19-CAR T-cell product (after treatment middle black and grey bars, FIG. 13B ).
  • mice were then challenged with a second inoculum of Raji tumor cells and the frequency of CD4+ and CD8+ CAR T cells in the peripheral blood, and tumor engraftment were analyzed.
  • mice treated with a combined CD8+ and CD4+ CAR T-cell product significantly higher levels CD8+ CAR T cells after the tumor challenge ( FIG. 13C , lower panels), and complete rejection of the Raji inoculum (after tumor challenge right grey bar, FIG. 13B ).
  • mice that had received CD8+ CD19-CAR CTL alone we did not detect an increase in CAR T cells after the tumor challenge ( FIG. 13C ) and the Raji tumor cells were able to engraft (after tumor challenge right black bar, panel FIG. 13B ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Wood Science & Technology (AREA)
  • Communicable Diseases (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention provides methods and compositions to confer and/or augment immune responses mediated by cellular immunotherapy, such as by adoptively transferring genetically modified tumor specific CD8+ T cells in the presence of tumor-specific, subset specific genetically modified CD4+ T cells, wherein the CD4+ T cells confer and/or augment a CD8+ T cells ability to sustain anti-tumor reactivity and increase and/or maximize tumor-specific proliferation of the tumor-specific CD8+ T cells of interest. Pharmaceutical formulations produced by the method, and methods of using the same, are also described.

Description

CROSS REFERENCE TO RELATED APPLICATIONS
The present application is a continuation of U.S. application Ser. No. 14/006,641 filed May 5, 2014, now allowed, which is a U.S. national phase application of PCT/US2012/030388 filed Mar. 23, 2012, which claims benefit of U.S. Provisional Application No. 61/466,552 filed Mar. 23, 2011 are herein incorporated by reference in their entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
This invention was made with government support under CA018029 awarded by the National Institutes of Health. The government has certain rights in the invention.
FIELD OF THE INVENTION
The present invention relates to the field of biomedicine and specifically methods useful for cancer therapy. In particular, embodiments of the invention relate to methods and compositions for carrying out cellular immunotherapy.
BACKGROUND OF THE INVENTION
Studies in rodents have demonstrated that adoptive immunotherapy with antigen specific T cells is effective for cancer and infections, and there is evidence this modality has therapeutic activity in humans1-8. For clinical applications, it is necessary to isolate T cells of a desired antigen specificity or to engineer T cells to express receptors that target infected or transformed cells, and then expand these cells in culture9-14. The transfer of T cell clones is appealing because it enables control of specificity and function, and facilitates evaluation of in vivo persistence, toxicity and efficacy. Additionally, in the setting of allogeneic stem cell transplantation, the administration to recipients of T cell clones from the donor that target pathogens or malignant cells can avoid graft-versus-host disease that occurs with infusion of unselected donor T cells3,4,15. However, it is apparent from clinical studies that the efficacy of cultured T cells, particularly cloned CD8+ T cells, is frequently limited by their failure to persist after adoptive transfer16,17.
The pool of lymphocytes from which T cells for adoptive immunotherapy can be derived contains naïve and long-lived, antigen experienced memory T cells (TM). TM can be divided further into subsets of central memory (TCM) and effector memory (TEM) cells that differ in phenotype, homing properties and function18. CD8+ TCM express CD62L and CCR7 at the cell surface, which promote migration into lymph nodes, and proliferate rapidly if re-exposed to antigen. CD8+ TEM lack cell surface CD62L and preferentially migrate to peripheral tissues, and exhibit immediate effector function19. In response to antigen stimulation, CD8+ TCM and TEM both differentiate into cytolytic effector T cells (TE) that express a high level of granzymes and perforin, but are short-lived20. Thus, the poor survival of T cells in clinical immunotherapy trials may simply result from their differentiation during in vitro culture to TE that are destined to die17,21,22. There is a need to identify cell populations and methods that provide enhanced survival of adoptively transferred T cells in vivo.
SUMMARY
In one aspect, the present invention relates to methods and compositions to confer and/or augment immune responses mediated by cellular immunotherapy, such as by adoptively transferring tumor-specific, subset specific genetically modified CD4+ T cells, wherein the CD4+ T cells confer and/or augment the ability of CD8+ T cells to sustain anti-tumor reactivity and increase and/or maximize tumor-specific proliferation.
In one embodiment, the present invention provides a method of performing cellular immunotherapy in a subject having a disease or disorder by administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation that provides a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as co-stimulatory domains; and a genetically modified helper T lymphocyte cell preparation that exhibits a predominant Th1 phenotype as well as produce other cytokines, elicits direct tumor recognition and augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor. Various modifications of the above method are possible. For example, the chimeric antigen receptor modifying the CD4+ T cell and the CD8+ T cell can be the same or different. In alternative embodiments, the T cells can be modified with a recombinant T cell receptor (TCR). TCR could be specific for any antigen, pathogen or tumor. There are TCRs for many tumor antigens in melanoma (MART1, gp100, for example), leukemia (WT1, minor histocompatibility antigens, for example), breast cancer (her2, NY-BR1, for example).
In another embodiment, the present invention provides an adoptive cellular immunotherapy composition having a genetically modified CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular variable domain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as a costimulatory domain, and a genetically modified helper T lymphocyte cell preparation that exhibits a predominant Th1 phenotype as well as produce other cytokines, elicits direct tumor recognition and augments the ability of genetically modified cytotoxic T lymphocyte cell preparations to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation has CD4+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In yet another embodiment, the present invention provides an adoptive cellular immunotherapy composition having a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and an antigen-reactive chimeric antigen receptor modified naïve CD4+ T helper cell that is derived from CD45RO negative, CD62L positive CD4 positive T cells, and a pharmaceutically acceptable carrier.
In another embodiment, the present invention provides an adoptive cellular immunotherapy composition having an antigen specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response comprising CD8+ T cells derived from the patient together with an antigen-reactive chimeric antigen receptor modified CD4+ T helper cell that elicits a Th1 cytokine response and augments the CD8+ immune response to pathogens, wherein the helper T lymphocyte cell preparation with CD4+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In another embodiment, the present invention provides an adoptive cellular immunotherapy composition with an antigen-reactive chimeric antigen receptor modified CD4+ T helper cell that elicits direct tumor recognition and augments the CD8+ immune response to pathogens, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with a disease or disorder and an intracellular signaling domain of a T cell receptor.
In another aspect, the present invention provides a method of manufacturing an adoptive immunotherapy composition by obtaining a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response and an antigen-reactive chimeric antigen receptor, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor; and obtaining a modified naïve CD4+ T helper cell that elicits a Th1 cytokine response, wherein the modified helper T lymphocyte cell preparation comprises CD4+ cells that have a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In another embodiment, the present invention provides a method of manufacturing an adoptive immunotherapy composition by obtaining a modified naïve CD4+ T helper cell that elicits a Th1 cytokine response, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and combining the modified naïve CD4+ T helper cell with an antigen specific central memory CD8+ cytotoxic T lymphocyte cell preparation that has a chimeric antigen receptor with an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors.
In one embodiment, the present invention provides a method of performing cellular immunotherapy in subject having a disease or disorder by administering to the subject a genetically modified helper T lymphocyte cell preparation, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor.
These and other embodiments of the invention are described further in the accompanying specification, drawings and claims.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows the phenotype and analysis of chimeric antigen receptor (CAR) expression in a CAR-transduced with ROR1-CAR encoding lentivirus, and an untransduced CD8+ T cell line as a control. The ROR1-CAR cassette contains a truncated EGFR that serves as transduction marker and can be detected by staining with anti-EGFR monoclonal antibodies. Truncated Fc-ROR1 fusion protein binds directly to the antigen-binding domain of the ROR1-CAR and selectively stains the ROR1-CAR transduced but not the untransduced control T cell line. Expression of the ROR1-CAR on the cell surface of CD8+ T cells is measured directly by binding to ROR1-Fc fusion protein and indirectly by expression of a truncated EGFR that is encoded downstream of a 2A sequence in the vector.
FIG. 2 shows cytolytic activity of CD8+ T cells expressing a ROR1-specific chimeric antigen receptor against a panel of human ROR1-positive tumor cell lines (K562) and primary tumor cells (B-CLL) and autologous normal B-cells in a 51Cr release assay. Consistent with the uniform expression of ROR1 on malignant but not on mature normal B cells, genetically modified CD8+ ROR1-CAR T cells only lysed ROR1+ tumor cells but not mature normal B cells. CD8+ ROR1-CAR T cells exert specific lytic activity against ROR1-positive tumor cells including primary CLL, but not against normal B cells.
FIG. 3 shows the phenotype and CAR expression of a ROR1-CAR transduced and an untransduced CD4+ T cell line as a control. Expression of the ROR1-CAR on the cell surface of CD4+ T cells is measured by specific binding to ROR1-Fc fusion protein. Truncated Fc ROR1 fusion protein but not Fc protein alone binds directly to the ROR1-CAR and selectively stains the ROR1-CAR transduced but not the untransduced control CD4+ T cell line confirming expression of the ROR1-CAR on the cell surface and binding to ROR1-protein. Expression of the ROR1-CAR on the cell surface of CD4+ T cells is measured by specific binding to ROR1-Fc fusion protein, but not to a control Fc fusion protein.
FIGS. 4A and 4B show weak but specific cytolytic activity of CD4+ ROR1-CAR T cells in a 51Cr release assay against a panel of ROR1-positive tumor cells including primary CLL, the mantle cell lymphoma line Jeko-1, K562 cells that were stably transfected with ROR1 (K562/ROR1), but not native ROR1-negative K562 cells. CD4+ ROR1-CAR T cells exert weak but specific lytic activity against ROR1-positive tumor cells.
FIGS. 5A and 5B show the results from an IFNγ ELISA (FIG. 5A) and multiplex cytokine assay (FIG. 5B). Cytokine secretion of CD4+ and CD8+ ROR1-CAR T cell lines. CD4+ROR1-CAR and CD8 ROR1-CAR T cells were co-incubated with ROR1+ tumor cells, and levels of interferon gamma (IFNγ) was measured by ELISA (5A), and IFNγ, TNFα, IL-2, IL-4, IL-10 and IL-17 were measured by Luminex assay (5B). CD4+ ROR1-CAR modified T cells specifically recognize ROR1-positive tumor cells and tumor cell lines and produce higher amounts of Th1 cytokines including IFN-γ, TNF-α and particularly IL-2 than CD8+ ROR1-CAR modified T cells. These data demonstrate that CD4+ ROR1-CAR T cells exert helper effector functions after stimulation through the ROR1-CAR and in addition to mediating direct anti-tumor reactivity, could also be utilized to augment the ability of CD8+ ROR1-CAR modified T cells to mediate a cellular immune response.
FIG. 6 depicts the results of a proliferation study showing that CD4+ ROR1-CAR T cells are induced to proliferate after stimulation with ROR1-positive tumor cell lines and primary tumor cells (CFSE assay) and that both the percentage of proliferating cells and number of cell divisions that the proliferating subset underwent were significantly higher compared to CD8+ ROR1-CAR modified T cells. CD4+ ROR1-CAR T cells proliferate more vigorously after stimulation with ROR1-positive tumor cells (K562/ROR1, primary CLL, and Jeko MCL) compared to CD8+ ROR1-CAR CTLs.
FIG. 7 shows polyclonal unselected CD4+ ROR1 CAR T cells provide help to CD8+ ROR1-CAR CTLs by promoting their proliferation in response to tumor. CD4+ ROR1-CAR T cells (derived from bulk CD4+ T cells) significantly increased proliferation of polyclonal unselected CD8+ ROR1-CAR CTLs (18% in individual culture→31.5% after co-culture with CD4+ CAR T cells).
FIGS. 8A-8D show the generation of CD4+ CAR T cell lines from flow sort purified CD4+ naïve, central memory and effector memory subsets and analysis of T-cell function. Cytokine profile and proliferative capacity suggest that CD4+ ROR1-CAR T cells derived from naïve CD4+ T cells may be best suited to provide help to CD8+ CTLs. Similar data were obtained in experiments comparing the function of CD4+ CAR T-cell lines expressing a CD19-specific CAR. FIG. 8A shows flow sort purification of naïve, central and effector memory CD4+ T cells based on expression of CD45RA, CD45RO, CD62L. FIG. 8B shows analysis of proliferation of ROR1-CAR T cell lines that were derived by lentiviral transduction of sort purified naïve, central and effector memory CD4+ T cells (CFSE assay). FIG. 8C shows analysis of cytokine secretion of ROR1-CAR T cell lines from sort purified naïve, central and effector memory CD4+ T cells (Luminex assay). FIG. 8D shows analysis of cytokine secretion of CD19-CAR T-cell lines from sort purified naïve, central and effector memory CD4+ T cells (Luminex assay). The cytokine profile obtained by multiplex cytokine analysis (FIG. 8B) and proliferative capacity by CFSE staining (FIG. 8C) shows that CD4+ ROR1-CAR modified T cells derived from the naïve subset produced the highest levels of Th1 cytokines and proliferated most vigorously after stimulation with ROR1-positive tumor cells, suggesting they may be best suited to augment CD8+ ROR1-CAR CTLs. Analysis of cytokine secretion of CD19-CAR T cell lines from sort-purified naïve, central and effector memory CD4+ T cells (Luminex assay), demonstrates that the activity of CD4 T cell subsets is generalizable to many CARs.
FIG. 9 shows co-culture of CD8+ ROR1-CAR modified T cells with CD4+ ROR1-CAR modified T cells (but not untransduced control CD4+ T cells). Co-culture of CD8+ ROR1-CAR CTLs and CD4+ ROR1-CAR T cell lines derived from naïve, central and effector memory subsets to define the optimal combination of CD8+ and CD4+ T cells that would allow maximum proliferation of CD8+ ROR1-CAR CTLs. CD4 naïve ROR1-CAR T cells provide the greatest proliferation of CD8 central memory ROR1-CAR CTLs. Co culture leads to an increase in tumor-specific proliferation of the CD8+ subset, and that maximum proliferation of the CD8+ subset is observed after co-culture with CD4+ ROR1-CAR T cells derived from naïve CD4+ T cells.
FIG. 10 shows the superior ability of CD4+ CAR T-cell lines derived from the naïve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1. The superior ability of CD4+ CAR T-cell lines derived from the naïve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL was confirmed in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1.
FIGS. 11A and 11B show that CD8+ CAR T cells and CD4+ CAR T cells independently confer direct anti-tumor efficacy in a lymphoma model in immunodeficient mice (NOD/SCID-Raji). Groups of mice (n=3) were inoculated with firefly-luciferase expressing Raji tumor cells via tail vein injection and treated with a single dose of 10×10{circumflex over ( )}6 T cells. Mice received either CD19-CAR transduced or control mock-transduced CD8+ central memory-derived (FIG. 11A), or CD19-CAR transduced or control mock-transduced CD4+ naïve-derived T cells (FIG. 11B). Tumor burden and distribution was analyzed using serial bioluminescence imaging.
FIG. 12 shows the augmentation and synergistic effect CD4+ ROR1-CAR modified T cells on the anti-tumor efficacy of CD8+ ROR1-CAR CTLs in a mouse tumor model of systemic mantle cell lymphoma (NSG/Jeko-1-ffLuc). Anti-tumor efficacy of ROR1-CAR modified CD8+ and CD4+ T cells in a mouse tumor model of systemic aggressive mantle cell lymphoma (NSG/Jeko-1). Analysis of tumor burden using bioluminescence imaging after adoptive transfer of CD8+ ROR1-CAR CTLs, CD4+ ROR1-CAR T cells or a combination of CD8+ and CD4+ ROR1-CAR T cells T cells. All mice received the same total dose of CAR T cells.
FIGS. 13A-13D show synergy of CD8+ and CD4+ CD19-CAR T cells in a mouse model of systemic lymphoma (NSG/Raji). NSG mice were inoculated with firefly-luciferase transduced Raji tumor cells. Engraftment of the Raji tumor was confirmed by bioluminescence imaging on day 6 after tumor inoculation (before treatment) (treatment scheme shown in FIG. 13A, tumor engraftment by bioluminescence shown in FIG. 13B. Groups of mice (n=5) were then treated with either CD8+ CD19-CAR modified T cells, or a combined T-cell product that contained both CD8+ and CD4+ CD19-CAR T cells. All mice received the same total dose of T cells (10×106). Analysis of tumor burden using bioluminescence imaging showed complete eradication of the Raji tumors in the cohorts of mice treated with CD8+ CD19-CAR T cells, and in mice treated with the combined CD8+ and CD4+ CD19-CAR T-cell product (after treatment middle black and grey bars) (FIG. 13B). The mice were then challenged with a second inoculum of Raji tumor cells and the frequency of CD4+ and CD8+ CAR T cells in the peripheral blood, and tumor engraftment were analyzed. In mice treated with a combined CD8+ and CD4+ CAR T-cell product, significantly higher levels CD8+ CAR T cells after the tumor challenge (FIG. 13C, lower panels), and complete rejection of the Raji inoculum (after tumor challenge right grey bar, FIG. 13B). In contrast, in mice that had received CD8+ CD19-CAR CTL alone, we did not detect an increase in CAR T cells after the tumor challenge (FIG. 13C) and the Raji tumor cells were able to engraft (after tumor challenge right black bar, panel FIG. 13B).
DETAILED DESCRIPTION
“T cells” or “T lymphocytes” as used herein may be from any mammalian, preferably primate, species, including monkeys, dogs, and humans. In some embodiments the T cells are allogeneic (from the same species but different donor) as the recipient subject; in some embodiments the T cells are autologous (the donor and the recipient are the same); in some embodiments the T cells arc syngeneic (the donor and the recipients are different but are identical twins).
Cytotoxic T lymphocyte (CTL) as used herein refers to a T lymphocyte that expresses CD8 on the surface thereof (i.e., a CD8+ T cell). In some embodiments such cells are preferably “memory” T cells (TM cells) that are antigen-experienced.
“Central memory” T cell (or “TCM”) as used herein refers to an antigen experienced CTL that expresses CD62L and CD45RO on the surface thereof, and does not express or has decreased expression of CD45RA as compared to naïve cells. In embodiments, central memory cells are positive for expression CD62L, CCR7, CD28, CD127, CD45RO, and CD95, and have decreased expression of CD54RA as compared to naïve cells.
“Effector memory” T cell (or “TEM”) as used herein refers to an antigen experienced CTL that does not express or has decreased expression of CD62L on the surface thereof as compared to central memory cells, and does not express or has decreased expression of CD45RA as compared to naïve cell. In embodiments, effector memory cells are negative for expression CD62L, CCR7, CD28, CD45RA, and are positive for CD127 as compared to naïve cells or central memory cells.
“Naïve” T cells as used herein refers to a non-antigen experienced T lymphocyte that expresses CD62L and CD45RA, and does not express or has decreased expression of CD45RO− as compared to central memory cells. In some embodiments, naïve CD8+ T lymphocytes are characterized by the expression of phenotypic markers of naïve T cells including CD62L, CCR7, CD28, CD3, CD127, and CD45RA.
“Effector” or “TE” T cells as used herein refers to a antigen experienced cytotoxic T lymphocyte cells that do not express or have decreased expression of CD62L, CCR7, CD28, and are positive for granzyme B and perforin as compared to central memory cells.
“Enriched” and “depleted” as used herein to describe amounts of cell types in a mixture refers to the subjecting of the mixture of the cells to a process or step which results in an increase in the number of the “enriched” type and a decrease in the number of the “depleted” cells. Thus, depending upon the source of the original population of cells subjected to the enriching process, a mixture or composition may contain 60, 70, 80, 90, 95, or 99 percent or more (in number or count) of the “enriched” cells and 40, 30, 20, 10, 5 or 1 percent or less (in number or count) of the “depleted” cells.
Interleukin-15 is a known and described in, for example, U.S. Pat. No. 6,344,192.
“CAR” as used herein refers to chimeric antigen receptor comprising an extracellular variable domain of an antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors, such as a costimulatory domain.
Modes of the Disclosure
CD4+ T lymphocytes during in vitro culture significantly increase proliferation, persistence and anti-tumor reactivity of tumor-specific CD8+ T cells in vitro and in vivo. In some embodiments, naïve CD4+ T cells possess an intrinsic programming that leads to superior helper activity compared to CD4+ T cells derived from central and effector memory, or bulk CD4+ T cells.
In embodiments, tumor-reactive CD4+ T cells are modified with a single-chain antibody-derived chimeric antigen receptor (CAR) specific for the orphan tyrosine kinase receptor ROR1 or for the CD19 molecule. ROR1 is uniformly expressed on chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) and ROR1-specific CAR from an anti-ROR1 monoclonal antibody (mAb) confers specific recognition of malignant, but not mature normal B-cells when expressed in CD8+ cytotoxic T cells (CTLs). ROR1-CAR T cells from bulk and flow sort purified naïve, central and effector memory CD4+ T cells are obtained from the peripheral blood of both healthy donors and CLL patients. CD4+ CAR T cells had specific but weak cytolytic activity against ROR1+ tumors including primary CLL, the MCL line Jeko-1, and K562 cells transfected with ROR1. Multiplex cytokine analysis detects high-level production of Th1 cytokines with significantly higher levels of IFNγ, TNFα, and particularly IL-2 compared to CD8+ CAR CTLs. CFSE staining shows dramatically higher proliferation after stimulation with ROR1-positive tumor cells, with both the percentage of cells that were induced to proliferate and the number of cell divisions that the proliferating subset underwent being significantly higher compared to CD8+ CAR CTL. CD4+ T cells obtained from both healthy donors and CLL patients acquire anti-tumor reactivity after genetic modification with a ROR1-specific CAR. Moreover, the ability to proliferate in the absence of exogenous cytokines and to produce high levels of Th1 cytokines demonstrates that CD4+ CAR T cells exert typical helper functions after stimulation through the CAR and suggests that in addition to conferring direct anti-tumor effects, could be utilized to augment tumor-specific CD8+ CTL.
The cytokine profile and proliferative capacity of ROR1-CAR T cells derived from flow sort purified CD4+ naïve, central and effector memory subsets is obtained. The CD4+ CAR T cells, derived from the naïve CD45RA+ CD45RO− CD62L+ subset, produces the highest levels of Th1 cytokines, especially IL-2, and proliferates in response to ROR1+ tumor cells. Indeed, in co-culture experiments, the addition of CAR-transduced, but not untransduced CD4+ T cells leads to a significant increase in tumor-specific proliferation of CD8+ CAR CTLs. In some embodiments, CAR-modified CD4+T cells derived from naïve rather than central and effector memory subsets or bulk CD4+ T cells results in enhanced proliferation of CD8+ CAR CTL.
CD8+ central memory T cells have an intrinsic programming that allows them to persist for extended periods after administration, which makes them the preferred subset of CD8+ T cells for immunotherapy. In embodiments, ROR1-CAR or CD19 CAR modified CTLs from sort purified CD8+ central memory T cells and CD4+ naïve CAR-modified T cells provide enhanced proliferation of the CD8+ T cell subset. In embodiments, tumor-specific CD4+ T cells exert anti-tumor reactivity and provide help to tumor-specific CD8+ T cells in vitro and in vivo. In a specific embodiment, tumor-specific CD4+ T cells from the naïve subset are utilized.
In another embodiment, the CD8+ and CD4+ T cells can be modified with a T cell receptor (TCR). The TCR could be specific for any antigen, pathogen or tumor (there are TCRs for many tumor antigens in melanoma (MART1, gp100 for example), leukemia (WT1, minor histocompatibility antigens for example), breast cancer (her2, NY-BR1 for example).
Compositions
The disclosure provides for an adoptive cellular immunotherapy composition comprising a genetically modified helper T lymphocyte cell preparation that augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor or other receptors.
In some embodiments, an adoptive cellular immunotherapy composition further comprises a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In some embodiments, an adoptive cellular immunotherapy composition comprises a chimeric antigen receptor modified tumor-specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular single chain antibody specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, in combination with an antigen-reactive chimeric antigen receptor modified naïve CD4+ T helper cell derived from CD45RO negative, CD62L positive CD4 positive T cells, and a pharmaceutically acceptable carrier.
In other embodiments, an adoptive cellular immunotherapy composition comprises an antigen specific CD8+ cytotoxic T lymphocyte cell preparation that elicits a cellular immune response derived from the patient combined with an antigen-reactive chimeric antigen receptor modified naïve CD4+ T helper cell that augments the CD8+ immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In a further embodiment, an adoptive cellular immunotherapy composition comprises an antigen-reactive chimeric antigen receptor modified naïve CD4+ T helper cell that augments the CD8+ immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with a disease or disorder and an intracellular signaling domain of a T cell receptor.
In embodiments, the CD4+ T helper lymphocyte cell is selected from the group consisting of naïve CD4+ T cells, central memory CD4+ T cells, effector memory CD4+ T cells, or bulk CD4+ T cells. In some embodiments, CD4+ helper lymphocyte cell is a naïve CD4+ T cell, wherein the naïve CD4+ T cell comprises a CD45RO−, CD45RA+, CD62L+ CD4+ T cell. In embodiments, the CD8+ T cytotoxic lymphocyte cell is selected from the group consisting of naïve CD8+ T cells, central memory CD8+ T cells, effector memory CD8+ T cells or bulk CD8+ T cells. In some embodiments, the CD8+ cytotoxic T lymphocyte cell is a central memory T cell wherein the central memory T cell comprises a CD45RO+, CD62L+, CD8+ T cell. In yet other embodiments, the CD8+ cytotoxic T lymphocyte cell is a central memory T cell and the CD4+ helper T lymphocyte cell is a naïve CD4+ T cell.
In alternative embodiments, the T cells can be modified with a recombinant T cell receptor. TCR could be specific for any antigen, pathogen or tumor. There are TCRs for many tumor antigens in melanoma (MART1, gp100, for example), leukemia (WT1, minor histocompatibility antigens, for example), breast cancer (her2, NY-BR1, for example).
Selection and Sorting of T Lymphocyte Populations
The compositions described herein provide for antigen reactive CD4+ and CD8+ T lymphocytes.
T lymphocytes can be collected in accordance with known techniques and enriched or depleted by known techniques such as affinity binding to antibodies such as flow cytometry and/or immunomagnetic selection. After enrichment and/or depletion steps, in vitro expansion of the desired T lymphocytes can be carried out in accordance with known techniques (including but not limited to those described in U.S. Pat. No. 6,040,177 to Riddell et al.), or variations thereof that will be apparent to those skilled in the art.
For example, the desired T cell population or subpopulation may be expanded by adding an initial T lymphocyte population to a culture medium in vitro, and then adding to the culture medium feeder cells, such as non-dividing peripheral blood mononuclear cells (PBMC) (e.g., such that the resulting population of cells contains at least about 5, 10, 20, or 40 or more PBMC feeder cells for each T lymphocyte in the initial population to be expanded); and incubating the culture (e.g., for a time sufficient to expand the numbers of T cells). The non-dividing feeder cells can comprise gamma-irradiated PBMC feeder cells. In some embodiments, the PBMC are irradiated with gamma rays in the range of about 3000 to 3600 rads. The order of addition of the T cells and feeder cells to the culture media can be reversed if desired. The culture can typically be incubated under conditions of temperature and the like that are suitable for the growth of T lymphocytes. For the growth of human T lymphocytes, for example, the temperature will generally be at least about 25 degrees Celsius, preferably at least about 30 degrees, more preferably about 37 degrees.
The T lymphocytes expanded include cytotoxic T lymphocytes (CTL) and helper T lymphocytes that are specific for an antigen present on a human tumor or a pathogen.
Optionally, the expansion method may further comprise the step of adding non-dividing EBV-transformed lymphoblastoid cells (LCL) as feeder cells. LCL can be irradiated with gamma rays in the range of about 6000 to 10,000 rads. The LCL feeder cells may be provided in any suitable amount, such as a ratio of LCL feeder cells to initial T lymphocytes of at least about 10:1.
Optionally, the expansion method may further comprise the step of adding anti-CD3 monoclonal antibody to the culture medium (e.g., at a concentration of at least about 0.5 ng/ml). Optionally, the expansion method may further comprise the step of adding IL-2 and/or IL-15 to the culture medium (e.g., wherein the concentration of IL-2 is at least about 10 units/ml).
After isolation of T lymphocytes both cytotoxic and helper T lymphocytes can be sorted into naïve, memory, and effector T cell subpopulations either before or after expansion.
CD8+ cells can be obtained by using standard methods. In some embodiments, CD8+ cells are further sorted into naïve, central memory, and effector cells by identifying cell surface antigens that are associated with each of those types of CD8+ cells. In embodiments, memory T cells are present in both CD62L+ and CD62L− subsets of CD8+ peripheral blood lymphocytes. PBMC are sorted into CD62L-CD8+ and CD62L+ CD8+ fractions after staining with anti-CD8 and anti-CD62L antibodies. In some embodiments, the expression of phenotypic markers of central memory TCM include CD45RO, CD62L, CCR7, CD28, CD3, and CD127 and are negative for granzyme B. In some embodiments, central memory T cells are CD45RO+, CD62L+, CD8+ T cells. In some embodiments, effector TE are negative for CD62L, CCR7, CD28, and CD127, and positive for granzyme B and perforin. In some embodiments, naïve CD8+ T lymphocytes are characterized by the expression of phenotypic markers of naïve T cells including CD62L, CCR7, CD28, CD3, CD127, and CD45RA.
Whether a cell or cell population is positive for a particular cell surface marker can be determined by flow cytometry using staining with a specific antibody for the surface marker and an isotype matched control antibody. A cell population negative for a marker refers to the absence of significant staining of the cell population with the specific antibody above the isotype control, positive refers to uniform staining of the cell population above the isotype control. In some embodiments, a decrease in expression of one or markers refers to loss of 1 log 10 in the mean fluorescence intensity and/or decrease of percentage of cells that exhibit the marker of at least 20% of the cells, 25% of-the cells, 30% of the cells, 35% of the cells, 40% of the cells, 45% of the cells, 50% of the cells, 55% of the cells, 60% of the cells, 65% of the cells, 70% of the cells, 75% of the cells, 80% of the cells, 85% of the cells, 90% of the cell, 95% of the cells, and 100% of the cells and any % between 20 and 100% when compared to a reference cell population. In some embodiments, a cell population positive for of one or markers refers to a percentage of cells that exhibit the marker of at least 50% of the cells, 55% of the cells, 60% of the cells, 65% of the cells, 70% of the cells, 75% of the cells, 80% of the cells, 85% of the cells, 90% of the cell, 95% of the cells, and 100% of the cells and any % between 50 and 100% when compared to a reference cell population.
CD4+ T helper cells are sorted into naïve, central memory, and effector cells by identifying cell populations that have cell surface antigens. CD4+ lymphocytes can be obtained by standard methods. In some embodiments, naïve CD4+ T lymphocytes are CD45RO−, CD45RA+, CD62L+ CD4+ T cell. In some embodiments, central memory CD4+ cells are CD62L positive and CD45RO positive. In some embodiments, effector CD4+ cells are CD62L and CD45RO negative.
Populations of CD4+ and CD8+ that are antigen specific can be obtained by stimulating naïve or antigen specific T lymphocytes with antigen. For example, antigen specific T cell clones can be generated to Cytomegalovirus antigens by isolating T cells from infected subjects and stimulating the cells in vitro with the same antigen. Naïve T cells may also be used. Any number of antigens from tumor cells, cancer cells, or infectious agents may be utilized. Examples of such antigens include HIV antigens, HCV antigens, HBV antigens, CMV antigens, parasitic antigens, and tumor antigens such as orphan tyrosine kinase receptor ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, and CEA. In some embodiments, the adoptive cellular immunotherapy compositions are useful in the treatment of a disease or disorder including a solid tumor, hematologic malignancy, melanoma, or infection with a virus.
Modification of T Lymphocyte Populations
In some embodiments it may be desired to introduce functional genes into the T cells to be used in immunotherapy in accordance with the present disclosure. For example, the introduced gene or genes may improve the efficacy of therapy by promoting the viability and/or function of transferred T cells; or they may provide a genetic marker to permit selection and/or evaluation of in vivo survival or migration; or they may incorporate functions that improve the safety of immunotherapy, for example, by making the cell susceptible to negative selection in vivo as described by Lupton S. D. et al., Mol. and Cell Biol., 11:6 (1991); and Riddell et al., Human Gene Therapy 3:319-338 (1992); see also the publications of PCT/US91/08442 and PCT/US94/05601 by Lupton et al. describing the use of bifunctional selectable fusion genes derived from fusing a dominant positive selectable marker with a negative selectable marker. This can be carried out in accordance with known techniques (see, e.g., U.S. Pat. No. 6,040,177 to Riddell et al. at columns 14-17) or variations thereof that will be apparent to those skilled in the art based upon the present disclosure.
In embodiments, T cells are modified with chimeric antigen receptors (CAR). In some embodiments, CARs comprise a single-chain antibody fragment (scFv) that is derived from the variable heavy (VH) and variable light (VL) chains of a monoclonal antibody (mAb) linked to the TCR CD3+ chain that mediates T-cell activation and cytotoxicity. Costimulatory signals can also be provided through the CAR by fusing the costimulatory domain of CD28 or 4-1BB to the CD3+ chain. CARs are specific for cell surface molecules independent from HLA, thus overcoming the limitations of TCR-recognition including HLA-restriction and low levels of HLA-expression on tumor cells.
CARs can be constructed with a specificity for any cell surface marker by utilizing antigen binding fragments or antibody variable domains of, for example, antibody molecules. The antigen binding molecules can be linked to one or more cell signaling modules. In embodiments, cell signaling modules include CD3 transmembrane domain, CD3 intracellular signaling domains, and CD 28 transmembrane domains. In embodiments, the intracellular signaling domain comprises a CD28 transmembrane and signaling domain linked to a CD3 intracellular domain. In some embodiments, a CAR can also include a transduction marker, such as tEGFR.
In embodiments, the intracellular signaling domain of the CD8+ cytotoxic T cells is the same as the intracellular signaling domain of the CD4+ helper T cells. In other embodiments, the intracellular signaling domain of the CD8+ cytotoxic T cells is different than the intracellular signaling domain of the CD4+ helper T cells.
In some embodiments, the CD8+ T cell and the CD4+ T cell are both genetically modified with an antibody heavy chain domain that specifically binds a pathogen-specific cell surface antigen. In embodiments, CARs are specific for cell surface expressed antigens associated with pathogens, tumors, or cancer cells. In some embodiments, a CAR is specific for HIV antigens, HCV antigens, HBV antigens, CMV antigens, parasitic antigens, and tumor antigens such as orphan tyrosine kinase receptor ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, and CEA. Methods for producing a CAR are described herein and can also be found in U.S. Pat. No. 6,410,319 by Forman and WO 2002/077029, U.S. Pat. No. 7,446,191, 2010/065818, 2010/025177, 2007/059298, and U.S. Pat. No. 7,514,537 by Jensen et al. and as described by Berger C. et al., J. Clinical Investigation, 118:1 294-308 (2008), which are hereby incorporated by reference.
In embodiments, the same or a different CAR can be introduced into each of CD4+ and CD8+ T lymphocytes. In embodiments, the CAR in each of these populations has an antigen binding molecule that specifically binds to the same antigen. The cellular signaling modules can differ. In embodiments each of the CD4 or CD8 T lymphocytes can be sorted in to naïve, central memory, effector memory or effector cells prior to transduction. In alternative embodiments, each of the CD4 or CD8 T lymphocytes can be sorted in to naïve, central memory, effector memory, or effector cells prior to transduction.
In alternative embodiments, the T cells can be modified with a recombinant T cell receptor. TCR could be specific for any antigen, pathogen or tumor. There are TCRs for many tumor antigens in melanoma (MART1, gp100 for example), leukemia (WT1, minor histocompatibility antigens for example), breast cancer (her2, NY-BR1 for example).
Various infection techniques have been developed which utilize recombinant infectious virus particles for gene delivery. This represents a currently preferred approach to the transduction of T lymphocytes of the present invention. The viral vectors which have been used in this way include virus vectors derived from simian virus 40, adenoviruses, adeno-associated virus (AAV), lentiviral vectors, and retroviruses. Thus, gene transfer and expression methods are numerous but essentially function to introduce and express genetic material in mammalian cells. Several of the above techniques have been used to transduce hematopoietic or lymphoid cells, including calcium phosphate transfection, protoplast fusion, electroporation, and infection with recombinant adenovirus, adeno-associated virus and retrovirus vectors. Primary T lymphocytes have been successfully transduced by electroporation and by retroviral infection.
Retroviral vectors provide a highly efficient method for gene transfer into eukaryotic cells. Moreover, retroviral integration takes place in a controlled fashion and results in the stable integration of one or a few copies of the new genetic information per cell.
It is contemplated that overexpression of a stimulatory factor (for example, a lymphokine or a cytokine) may be toxic to the treated individual. Therefore, it is within the scope of the invention to include gene segments that cause the T cells of the invention to be susceptible to negative selection in vivo. By “negative selection” is meant that the infused cell can be eliminated as a result of a change in the in vivo condition of the individual. The negative selectable phenotype may result from the insertion of a gene that confers sensitivity to an administered agent, for example, a compound. Negative selectable genes are known in the art, and include, inter alia the following: the Herpes simplex virus type I thymidine kinase (HSV-I TK) gene (Wigler et al., Cell 11:223, 1977) which confers ganciclovir sensitivity; the cellular hypoxanthine phosphribosyltransferase (HPRT) gene, the cellular adenine phosphoribosyltransferase (APRT) gene, bacterial cytosine deaminase, (Mullen et al., Proc. Natl. Acad. Sci. USA. 89:33, 1992).
In some embodiments it may be useful to include in the T cells a positive marker that enables the selection of cells of the negative selectable phenotype in vitro. The positive selectable marker may be a gene which, upon being introduced into the host cell expresses a dominant phenotype permitting positive selection of cells carrying the gene. Genes of this type are known in the art, and include, inter alia, hygromycin-B phosphotransferase gene (hph) which confers resistance to hygromycin B, the amino glycoside phosphotransferase gene (neo or aph) from Tn5 which codes for resistance to the antibiotic G418, the dihydrofolate reductase (DHFR) gene, the adenosine daminase gene (ADA), and the multi-drug resistance (MDR) gene.
Preferably, the positive selectable marker and the negative selectable element are linked such that loss of the negative selectable element necessarily also is accompanied by loss of the positive selectable marker. Even more preferably, the positive and negative selectable markers are fused so that loss of one obligatorily leads to loss of the other. An example of a fused polynucleotide that yields as an expression product a polypeptide that confers both the desired positive and negative selection features described above is a hygromycin phosphotransferase thymidine kinase fusion gene (HyTK). Expression of this gene yields a polypeptide that confers hygromycin B resistance for positive selection in vitro, and ganciclovir sensitivity for negative selection in vivo. See Lupton et al., Mol. Cell. Biol. 11:3374-3378, 1991. In addition, in preferred embodiments, the polynucleotides of the invention encoding the chimeric receptors are in retroviral vectors containing the fused gene, particularly those that confer hygromycin B resistance for positive selection in vitro, and ganciclovir sensitivity for negative selection in vivo, for example the HyTK retroviral vector described in Lupton, S. D. et al. (1991), supra. See also the publications of PCT/US91/08442 and PCT/US94/05601, by S. D. Lupton, describing the use of bifunctional selectable fusion genes derived from fusing a dominant positive selectable marker with negative selectable markers.
Preferred positive selectable markers are derived from genes selected from the group consisting of hph, nco, and gpt, and preferred negative selectable markers are derived from genes selected from the group consisting of cytosine deaminase, HSV-I TK, VZV TK, HPRT, APRT and gpt. Especially preferred markers are bifunctional selectable fusion genes wherein the positive selectable marker is derived from hph or neo, and the negative selectable marker is derived from cytosine deaminase or a TK gene or selectable marker.
A variety of methods can be employed for transducing T lymphocytes, as is well known in the art. For example, retroviral transductions can be carried out as follows: on day 1 after stimulation using REM as described herein, provide the cells with 20-30 units/ml IL-2; on day 3, replace one half of the medium with retroviral supernatant prepared according to standard methods and then supplement the cultures with 5 μg/ml polybrene and 20-30 units/ml IL-2; on day 4, wash the cells and place them in fresh culture medium supplemented with 20-30 units/ml IL-2; on day 5, repeat the exposure to retrovirus; on day 6, place the cells in selective medium (containing, e.g., an antibiotic corresponding to an antibiotic resistance gene provided in the retroviral vector) supplemented with 30 units/ml IL-2; on day 13, separate viable cells from dead cells using Ficoll Hypaque density gradient separation and then subclone the viable cells.
CD4+ and CD8+ cells can be modified with an expression vector encoding a CAR. In embodiments, these cells are then further sorted into subpopulations of naïve, central memory and effector cells as described above by sorting for cell surface antigens unique to each of those cell populations. In addition, CD4+ or CD8+ cell populations may be selected by their cytokine profile or proliferative activities. For example, CD4+ T lymphocytes that have enhanced production of cytokines such as IL-2, IL-4, IL-10, TNFα, and IFNγ as compared to sham transduced cells or transduced CD8+ cells when stimulated with antigen can be selected. In other embodiments, naïve CD4+ T cells that have enhanced production of IL-2 and/or TNFα are selected. Likewise, CD8+ cells that have enhanced IFNγ production are selected as compared to sham transduced CD8+ cells.
In embodiments, CD4+ and CD8+ cells that proliferate in response to antigen are selected. For example, CD4+ cells that proliferate vigorously when stimulated with antigen as compared to sham transduced cells, or CD8+ transduced cells are selected.
In some embodiments, CD4+ and CD8+ cells are selected that are cytotoxic for antigen bearing cells. In embodiments, CD4+ are expected to be weakly cytotoxic as compared to CD8+ cells.
The disclosure contemplates that combinations of CD4+ and CD8+ T cells will be utilized in the compositions. In one embodiment, combinations of CAR transduced CD4+ cells can be combined with CD8+ antigen reactive cells to the same antigenic specificity as the CAR. In other embodiments, CAR transduced CD8+ cells are combined with antigen reactive CD4+ cells. In yet another embodiment, CAR modified CD4+ and CD8+ cells are combined.
As described herein, the disclosure contemplates that CD4+ and CD8+ cells can be further separated into subpopulations, such as naïve, central memory, and effector cell populations. As described herein, in some embodiments, naïve CD4+ cells are CD45RO−, CD45RA+, CD62L+ CD4+ T cells. In some embodiments, central memory CD4+ cells are CD62L positive and CD45RO positive. In some embodiments, effector CD4+ cells are CD62L negative and CD45RO positive. Each of these populations may be independently modified with a CAR.
As described herein, in embodiments, memory T cells are present in both CD62L+ and CD62L− subsets of CD8+ peripheral blood lymphocytes. PBMCs are sorted into CD62L-CD8+ and CD62L+ CD8+ fractions after staining with anti-CD8 and anti-CD62L antibodies. In some embodiments, expression of phenotypic markers of central memory TCM include CD62L, CCR7, CD28, CD3, and CD127 and are negative for granzyme B. In some embodiments, central memory T cells are CD45RO+, CD62L+, CD8+ T cells. In some embodiments, effector TE cells are negative for CD62L, CCR7, CD28, and CD127, and positive for granzyme B and perforin. In some embodiments, naïve CD8+T lymphocytes are characterized by CD8+, CD62L+, CD45RO+, CCR7+, CD28+CD127+, and CD45RO+. Each of these populations may be independently modified with a CAR.
Each of the subpopulations of CD4+ and CD8+ cells can be combined with one another. In a specific embodiment, modified naïve CD4+ cells are combined with modified central memory CD8+ T cells to provide a synergistic cytotoxic effect on antigen bearing cells, such as tumor cells.
Methods
The disclosure provides methods of making adoptive immunotherapy compositions and uses or methods of using these compositions for performing cellular immunotherapy in a subject having a disease or disorder.
In embodiments, a method of manufacturing the compositions comprises obtaining a modified naïve CD4+ T helper cell, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain.
In another embodiment, a method further comprises obtaining a modified CD8+ cytotoxic T cell, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In another embodiment, a method comprises obtaining a modified CD8+ cytotoxic T cell, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor, and further comprising combining the modified CD8+ cytotoxic T cells with an antigen specific CD4+ helper cell lymphocyte cell preparation.
The preparation of the CD4+ and CD8+ cells that are modified with a CAR has been described above as well as in the examples. Antigen specific T lymphocytes can be obtained from a patient having the disease or disorder or can be prepared by in vitro stimulation of T lymphocytes in the presence of antigen. Subpopulations of CD4+ and CD8+ T lymphocytes can also be isolated as described herein and combined in the methods of manufacturing.
The disclosure also provides methods of performing cellular immunotherapy in a subject having a disease or disorder comprising: administering a composition. In other embodiments, a method comprises administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation that provides a cellular immune response, wherein the cytotoxic T lymphocyte cell preparation comprises CD8+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling domain of a T cell or other receptors and a genetically modified helper T lymphocyte cell preparation that elicits direct tumor recognition and augments the genetically modified cytotoxic T lymphocyte cell preparations ability to mediate a cellular immune response, wherein the helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising an extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling domain of a T cell receptor.
In another embodiment, a method of performing cellular immunotherapy in subject having a disease or disorder comprises: administering to the subject a genetically modified helper T lymphocyte cell preparation, wherein the modified helper T lymphocyte cell preparation comprises CD4+ T cells that have a chimeric antigen receptor comprising a extracellular antibody variable domain specific for an antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor. In an embodiments, the method further comprises administering to the subject a genetically modified cytotoxic T lymphocyte cell preparation, wherein the modified cytotoxic T lymphocyte cell preparation comprises CD8 positive cells that have a chimeric antigen receptor comprising a extracellular antibody variable domain specific for the antigen associated with the disease or disorder and an intracellular signaling module of a T cell receptor.
Another embodiment describes a method of performing cellular immunotherapy in a subject having a disease or disorder comprising: analyzing a biological sample of the subject for the presence of an antigen associated with the disease or disorder and administering the adoptive immunotherapy compositions described herein, wherein the chimeric antigen receptor specifically binds to the antigen.
A CAR is produced that has a component that provides for specific binding to an antigen associated with a disease or conditions, such as a solid tumor, cancer, viral infection, and an infection with a parasite. In embodiments, the intracellular signaling module of a T cell receptor of the chimeric antigen receptor comprises a transmembrane domain, a CD28 signaling domain, and a CD3 intracellular signaling domain, or other domains of T cell costimulatory molecules. In some embodiments, the intracellular signaling molecule comprises the CD3 intracellular domain, a CD28 domain, a CD28 transmembrane and signaling domain linked to a CD3 intracellular domain, or other domains of T cell costimulatory molecules.
In alternative embodiments, the T cells can be modified with a recombinant T cell receptor. TCR could be specific for any antigen, pathogen or tumor. There are TCRs for many tumor antigens in melanoma (MART1, gp100 for example), leukemia (WT1, minor histocompatibility antigens for example), breast cancer (her2, NY-BR1 for example).
In some embodiments, the CD4+ T helper lymphocyte cell is selected from the group consisting of naïve CD4+ T cells, central memory CD4+ T cells, effector memory CD4+ T cells or bulk CD4+ T cells. In a specific embodiment, CD4+ helper lymphocyte cell is a naïve CD4+ T cell, wherein the naïve CD4+ T cell comprises a CD45RO−, CD45RA+, CD62L+ CD4+ T cell. In yet other embodiments, the CD8+ T cytotoxic lymphocyte cell is selected from the group consisting of naïve CD8+ T cells, central memory CD8+ T cells, effector memory CD8+ T cells or bulk CD8+ T cells. In a specific embodiment, the CD8+ cytotoxic T lymphocyte cell is a central memory T cell wherein the central memory T cell comprises a CD45RO+, CD62L+, CD8+ T cell. In a specific embodiment, the CD8+ cytotoxic T lymphocyte cell is a central memory T cell and the CD4+ helper T lymphocyte cell is a naïve CD4+ T cell.
In embodiments, the CD8+ T cell and the CD4+ T cell are both genetically modified with a CAR comprising an antibody heavy chain domain that specifically binds a pathogen or tumor-specific cell surface antigen. In other embodiments, the intracellular signaling domain of the CD8 cytotoxic T cells is the same as the intracellular signaling domain of the CD4 helper T cells. In yet other embodiments, the intracellular signaling domain of the CD8 cytotoxic T cells is different than the intracellular signaling domain of the CD4 helper T cells.
Subjects that can be treated by the present invention are, in general, human and other primate subjects, such as monkeys and apes for veterinary medicine purposes. The subjects can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects.
The methods are useful in the treatment of, for example, solid tumor, hematologic malignancy, melanoma, or infection with a virus or other pathogen. Infections with pathogens include HIV, HCV, HBV, CMV, and parasitic disease. In some embodiments, the antigen associated with the disease or disorder is selected from the group consisting of orphan tyrosine kinase receptor ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, CEA, and hepatitis B surface antigen.
Subjects that can be treated include subjects afflicted with cancer, including but not limited to colon, lung, liver, breast, prostate, ovarian, skin (including melanoma), bone, and brain cancer, etc. In some embodiments the tumor associated antigens are known, such as melanoma, breast cancer, squamous cell carcinoma, colon cancer, leukemia, myeloma, prostate cancer, etc. (in these embodiments memory T cells can be isolated or engineered by introducing the T cell receptor genes). In other embodiments the tumor associated proteins can be targeted with genetically modified T cells expressing an engineered immunoreceptor. Examples include but are not limited to B cell lymphoma, breast cancer, prostate cancer, and leukemia.
Subjects that can be treated also include subjects afflicted with, or at risk of developing, an infectious disease, including but not limited to viral, retroviral, bacterial, and protozoal infections, etc. Subjects that can be treated include immunodeficient patients afflicted with a viral infection, including but not limited to Cytomegalovirus (CMV), Epstein-Barr virus (EBV), adenovirus, BK polyomavirus infections in transplant patients, etc.
Cells prepared as described above can be utilized in methods and compositions for adoptive immunotherapy in accordance with known techniques, or variations thereof that will be apparent to those skilled in the art based on the instant disclosure. See, e.g., US Patent Application Publication No. 2003/0170238 to Gruenberg et al; see also U.S. Pat. No. 4,690,915 to Rosenberg.
In some embodiments, the cells are formulated by first harvesting them from their culture medium, and then washing and concentrating the cells in a medium and container system suitable for administration (a “pharmaceutically acceptable” carrier) in a treatment-effective amount. Suitable infusion medium can be any isotonic medium formulation, typically normal saline, Normosol R (Abbott) or Plasma-Lyte A (Baxter), but also 5% dextrose in water or Ringer's lactate can be utilized. The infusion medium can be supplemented with human serum albumin.
A treatment-effective amount of cells in the composition is at least 2 cells (for example, 1 CD8+ central memory T cell and 1 CD4+ helper T cell subset) or is more typically greater than 102 cells, and up to 106, up to and including 108 or 109 cells and can be more than 1010 cells. The number of cells will depend upon the ultimate use for which the composition is intended as will the type of cells included therein. For example, if cells that are specific for a particular antigen are desired, then the population will contain greater than 70%, generally greater than 80%, 85% and 90-95% of such cells. For uses provided herein, the cells are generally in a volume of a liter or less, can be 500 mls or less, even 250 mls or 100 mls or less. Hence the density of the desired cells is typically greater than 106 cells/ml and generally is greater than 107 cells/ml, generally 108 cells/ml or greater. The clinically relevant number of immune cells can be apportioned into multiple infusions that cumulatively equal or exceed 109, 1010 or 1011 cells.
In some embodiments, the lymphocytes of the invention may be used to confer immunity to individuals. By “immunity” is meant a lessening of one or more physical symptoms associated with a response to infection by a pathogen, or to a tumor, to which the lymphocyte response is directed. The amount of cells administered is usually in the range present in normal individuals with immunity to the pathogen. Thus, the cells are usually administered by infusion, with each infusion in a range of from 2 cells, up to at least 106 to 1010 cells/m2, preferably in the range of at least 107 to 109 cells/m2. The clones may be administered by a single infusion, or by multiple infusions over a range of time. However, since different individuals are expected to vary in responsiveness, the type and amount of cells infused, as well as the number of infusions and the time range over which multiple infusions are given are determined by the attending physician, and can be determined by routine examination. The generation of sufficient levels of T lymphocytes (including cytotoxic T lymphocytes and/or helper T lymphocytes) is readily achievable using the rapid expansion method of the present invention, as exemplified herein. See, e.g., U.S. Pat. No. 6,040,177 to Riddell et al. at column 17.
The present invention is illustrated further in the examples sot forth below.
EXPERIMENTAL Example 1—T Cell Transduction and Analysis of CAR Expression
A ROR1-specific CAR can be expressed in human CD8+ T cells and confers specific recognition of ROR1+ B-cell tumors and not mature normal B cells. We constructed a ROR1-specific chimeric antigen receptor that when expressed in T cells from healthy donors or CLL patients conferred specific recognition of primary B-CLL and mantle cell lymphoma.
Materials and Methods
Cell Lines
Epstein-Barr virus transformed B cells (EBV-LCL) were generated as described (25). The tumor cell lines Jeko-1, and, BALL-1, were provided by Drs Oliver Press and Jerald Radich (Fred Hutchinson Cancer Research Center). All cell lines were maintained in RPMI, 10% fetal calf serum, 0.8 mM L-glutamine, and 1% penicillin-streptomycin (LCL medium). K562 cells were obtained from the American Type Culture Collection.
Transfection of K562 Cells with ROR1
For polymerase chain reaction (PCR)-amplification of the ROR1-gene, total RNA was obtained from B-CLL cells (RNeasyPlusKit; QIAGEN) and reverse transcribed into cDNA with M-MLVReverse Transcriptase (Invitrogen). PCR was performed with specific primers (ROR1-F: 5-XhoIAGAGGAGGAATGCACCGGCC-3 and ROR1-R: 5-XhoI-CACAGAAGGTACTTGTTGCGATGT-3) using Herculase-II DNA Polymerase (Stratagene). The PCR product was cloned into the MIGR-1 retroviral vector (23) and the sequence verified. Effectene transfection reagent (QIAGEN) was used to transfect Platinum-A cells (Cell Biolabs) with MIGR-1/ROR1 and produce ROR1-encoding retrovirus. K562 cells were retrovirally transduced by centrifugation at 2500 rpm for 60 minutes at 32° C., expanded, and the ROR1-positive subset was sort-purified.
Real-Time Quantitative PCR
First-strand cDNA of B-CLL, normal resting and activated B cells, and EBV-LCL was prepared as described in the previous paragraph. First-strand cDNA from normal tissues (Human Tissue panels I/II, Blood Fractions) was obtained from Clontech. Expression of ROR1 mRNA was analyzed in duplicate and normalized to GAPDH. Amplifications were performed on an ABI Prism 7900 (Applied Biosystems) in a 50 reaction consisting of 25 μL Power SYBR Green PCR Master Mix (Applied Biosystems), 2.5 ng of cDNA, and 300 nM gene-specific forward and reverse primers:
ROR1-F
5-AGCGTGCGATTCAAAGGATT-3,
ROR1-R
5-GACTGGTGCCGACGATGACT-3,
GAPDH-F
5-GAAGGTGAAGGTCGGAGTC-3,
and
GAPDH-R 
5-GAAGATGGTGATGGGATTTC-3.
The cycle threshold (Ct) was determined using SDS software v2.2.2 (Applied Biosystems) and the level of gene expression calculated using the comparative Ct method (2-(ΔΔCt)).
Vector Construction and Generation of Lentivirus
CD20-CAR (CD20R-epHIV7) and green fluorescent protein (GFP)-encoding lentiviral vectors (GFP-epHIV7) were described previously (24). The ROR1-CAR was encoded in the same vector. A mouse mAb (clone 2A2) that demonstrated specific binding to human ROR1 expressed on primary B-CLL and MCL tumor lines was generated, cloned, and characterized in a previous study. A codon-optimized nucleotide sequence encoding a scFv containing the VL and VH chain of mAb 2A2 was synthesized (GENEART) and cloned into CD20R-epHIV7 using NheI and RsrII restriction sites to replace the CD20-specific scFv. Lentivirus was produced in 293T cells co-transfected with the lentiviral vector and the packaging vectors pCHGP-2, pCMVRev2, and pCMV-G using Effectene (Qiagen). Medium was changed 16 hours after transfection and lentivirus collected after 48 hours.
Lentiviral Transduction and Isolation of CAR-Transduced Tcell Clones
PBMC from healthy donors and B-CLL patients, and sort-purified CD8+ CD45RO+ CD62L+ central memory T cells (TCM) were activated with anti-CD3 mAb (30 ng/mL) (25), and transduced in lentiviral supernatant supplemented with 1 μg/mL polybrene (Sigma-Aldrich) and 50 IU/mL recombinant human interleukin-2 (IL-2) on day 2 and 3 after activation by centrifugation at 2500 rpm for 60 minutes at 32° C. T cells were expanded in RPMI containing 10% human serum, 2 mM L-glutamine, and 1% penicillin streptomycin (CTL medium) (25). After expansion, an aliquot of each transduced T-cell line was stained with biotin-conjugated anti-EGFR (epithelial growth factor receptor) mAb, streptavidin-PE, and anti-CD8 mAb. EGFR+CD8+ T cells were sort purified and cloned by limiting dilution (0.5 cells/well) (25). ROR1-CAR transduced T cells were identified by staining with biotinylated recombinant Fc-ROR1 extracellular domain fusion protein and streptavidin-PE. Recombinant ROR1-protein was produced in transiently transfected 293F cells (Invitrogen), purified as described (26), and biotinylated using the BiotinTag kit (Sigma). GFP-transduced CD8+ T cells were identified by flow cytometry, sort-purified, and cloned in similar fashion.
Chromium Release and Cytokine Secretion Assays
Target cells were labeled with 51Cr (PerkinElmer) overnight, washed and incubated in triplicate at 1-2×103 cells/well with effector T cells at various effector to target (E:T) ratios. Supernatants were harvested for γ counting after a 4-hour incubation, and specific lysis was calculated using the standard formula (25).
Results
Transduced CD8+ T cells were sort-purified using a biotinylated anti-EGFR mAb and streptavidin conjugated dyes. ROR1-CAR expression on the surface of the sort-purified T cells was evaluated by staining the cells with a biotinylated recombinant Fc-ROR1 extracellular domain fusion protein that directly binds to the scFv of the ROR1-CAR, and costaining with streptavidin-conjugates. Fc-ROR1-protein specifically stained CD8+ T cells transduced with the ROR1-CAR lentiviral vector but not CD8+ T cells transduced with a control lentiviral vector encoding GFP (FIG. 1).
We established ROR1-CAR transduced (n=10) and control GFP-transduced CD8+ T-cell clones (n=4) by limiting dilution and confirmed the stable surface expression of the CAR after multiple rounds of in vitro expansion. There was no apparent difference in the growth of ROR1-CAR transduced compared with untransduced or GFP-transduced T-cell clones (data not shown).
The ROR1-CAR transduced T-cell clones efficiently lysed primary B-CLL and K562 cells that were stably transfected with the ROR1-gene, but not native, ROR1-negative K562 cells, demonstrating specific recognition of ROR1 (FIG. 2).
Discussion
Adoptive immunotherapies that employ CAR-modified T cells are being investigated in clinical trials for B-cell malignancies. The surface molecules that are being targeted are B-cell lineage-specific and include CD19, which is expressed on normal B-lineage cells from the pro-B-cell stage to plasma cells, and CD20, which is expressed on normal B cells from the pre-B-cell stage to memory B cells. Thus, an anticipated outcome of effective therapy targeting these molecules is depletion of normal B cells and B-cell precursors. Gene expression profiling studies have identified genes that are preferentially or exclusively expressed by malignant but not by normal B cells and ROR1 emerged as a CLL signature gene in 2 independent analyses (27,28). Specific antibodies to ROR1 developed in CLL patients after vaccination with autologous tumor cells that had been modified to express CD154 and treatment with lenalidomide without apparent toxicity to normal tissues, suggesting this tumor antigen may be a suitable target for immunotherapy (29,30).
Our studies illustrate the potential to target ROR1-positive malignant cells with engineered T cells expressing a ROR1-CAR. CD8+ ROR1-CAR T cells could be derived from both normal donors and CLL patients after lentiviral transduction of either bulk PBMCs or sort-purified TCM, that in animal models persist for extended periods after adoptive transfer (31). ROR1-CAR transduced T cells efficiently lysed primary B-CLL, but not normal resting or activated B-cells. These T cells produced effector cytokines including TNF-α, IFNγ, and IL-2, and were capable of proliferating in response to ROR1-expressing tumor cells.
Example 2—Generation of CD4+ CAR T Cell Lines and Analysis of Effector Function
CD4+ ROR1-CAR T cells can be generated from PBMC of healthy donors/CLL-patients. A ROR1-specific CAR can be expressed in human CD4+ T cells and confers specific recognition of ROR1+ B-cell tumors but not mature normal B cells.
Materials and Methods
Cell Lines
Epstein-Barr virus transformed B cells (EBV-LCL) were generated as described (25). The tumor cell lines Jeko-1, and BALL-1 were provided by Drs Oliver Press and Jerald Radich (Fred Hutchinson Cancer Research Center). All cell lines were maintained in RPMI, 10% fetal calf serum, 0.8 mM L-glutamine, and 1% penicillin-streptomycin (LCL medium). K562 and 293T cells were obtained from the American Type Culture Collection and cultured as directed.
Transfection of K562 Cells with ROR1
For polymerase chain reaction (PCR)-amplification of the ROR1-gene, total RNA was obtained from B-CLL cells (RNeasyPlusKit; QIAGEN) and reverse transcribed into cDNA with M-MLVReverse Transcriptase (Invitrogen). PCR was performed with specific primers (ROR1-F: 5-XhoIAGAGGAGGAATGCACCGGCC-3 and ROR1-R: 5-XhoI-CACAGAAGGTACTTGTTGCGATGT-3) using Herculase-II DNA Polymerase (Stratagene). The PCR product was cloned into the MIGR-1 retroviral vector (23), and sequence verified. Effectene transfection reagent (QIAGEN) was used to transfect Platinum-A cells (Cell Biolabs) with MIGR-1/ROR1 and produce ROR1-encoding retrovirus. K562 cells were retrovirally transduced by centrifugation at 2500 rpm for 60 minutes at 32° C., expanded, and the ROR1-positive subset was sort-purified.
Vector Construction and Generation of Lentivirus
CD20-CAR (CD20R-epHIV7) and green fluorescent protein (GFP)-encoding lentiviral vectors (GFP-epHIV7) were described previously (24). The ROR1-CAR was encoded in the same vector. A mouse mAb (clone 2A2) that demonstrated specific binding to human ROR1 expressed on primary B-CLL and MCL tumor lines was generated, cloned, and characterized in a previous study. A codon-optimized nucleotide sequence encoding a scFv containing the VL and VH chain of mAb 2A2 was synthesized (GENEART) and cloned into CD20R-epHIV7 using NheI and RsrII restriction sites to replace the CD20-specific scFv. Lentivirus was produced in 293T cells cotransfected with the lentiviral vector and the packaging vectors pCHGP-2, pCMVRev2, and pCMV-G using Effectene (Qiagen). Medium was changed 16 hours after transfection and lentivirus collected after 48 hours.
Lentiviral Transduction and Isolation of CD4+ ROR1-CAR T Cell Lines
CD4+ T cells were isolated from PBMC of healthy donors and activated with anti-CD3 mAb (30 ng/mL) (25), and transduced in lentiviral supernatant supplemented with 1 μg/mL polybrene (Sigma-Aldrich) and 50 IU/mL recombinant human interleukin-2 (IL-2) on day 2 and 3 after activation by centrifugation at 2500 rpm for 60 minutes at 32° C. T cells were expanded in RPMI containing 10% human serum, 2 mM L-glutamine, and 1% penicillin streptomycin (CTL medium).(25) After expansion, an aliquot of each transduced T-cell line was stained with biotin-conjugated anti-EGFR (epithelial growth factor receptor) mAb, streptavidin-PE, and anti-CD4 mAb. EGFR+ CD4+ T cells were sort purified and expanded. ROR1-CAR transduced T cells were identified by staining with biotinylated recombinant Fc-ROR1 extracellular domain fusion protein and streptavidin-PE. Recombinant ROR1-protein was produced in transiently transfected 293 cells (Invitrogen), purified as described (26), and biotinylated using the BiotinTag kit (Sigma). GFP-transduced CD4+ T cells were identified by flow cytometry, sort-purified, and cloned in similar fashion.
Chromium Release and Cytokine Secretion Assays
Target cells were labeled with 51Cr (PerkinElmer) overnight, washed and incubated in triplicate at 1-2×103 cells/well with effector T cells at various effector to target (E:T) ratios. Supernatants were harvested for γ counting after a 4-hour incubation, and specific lysis was calculated using the standard formula (25).
For analysis of cytokine secretion, target and effector cells were plated in triplicate wells at an E/T ratio of 2:1, and interferon IFNγ, tumor necrosis factor (TNF-α), and IL-2 were measured by multiplex cytokine immunoassay (Luminex) in supernatant removed after a 24-hour incubation.
CFSE Proliferation Assay
T cells were labeled with 0.2 μM carboxyfluorescein succinimidyl ester (CFSE; Invitrogen), washed, and plated with stimulator cells at a ratio of 2:1 in CTL medium containing 10 U/mL recombinant human IL-2. After a 72-hour incubation, cells were labeled with anti-CD4 mAb and propidium iodide (PI) to exclude dead cells from analysis. Samples were analyzed by flow cytometry, and cell division of live CD4+ T cells assessed by CFSE dilution.
Co-Culture Assay
ROR1-CAR transduced CD4+ T cells and ROR1-CAR transduced CD8+ cytotoxic T lymphocytes were labeled with CFSE, and co-cultured at a 2:1, 1:1 and 1:2 ratio. The co-cultures were then stimulated with K562/ROR1 cells and control K562 cells and cell proliferation measured by CFSE dye dilution assay after 5 days of incubation. For flow analysis, samples were stained with conjugated anti-CD8 and anti-CD4 mAb to distinguish CD8+ and CD4+ subsets.
Results
Generation of CD4+ ROR1-CAR T cells from PBMC of healthy donors and CLL patients We have shown that ROR1, an oncofetal tyrosine kinase receptor, is uniformly expressed on CLL and MCL, and developed a ROR1-CAR from an anti-ROR1 mAb that confers specific recognition of malignant, but not mature normal B cells when expressed in CD8+ T cells (32). Here, we generated CD4+ ROR1-CAR T cells to analyze direct tumor recognition and their ability to augment CD8+ ROR1-CAR CTL. CAR-modified CD4+ T cells could be readily generated from bulk peripheral CD4+ T cells of healthy donors (n=4) and CLL patients (n=4) using a ROR1-CAR encoding lentiviral vector. In this vector, we encoded a truncated EGFR (epithelial growth factor receptor, tEGFR) domain downstream of the ROR1-CAR and a self-cleavable 2A element, to serve both as transduction marker and for the enrichment of transgene expressing T cells with anti-EGFR mAb (FIG. 3). We determined the frequency of CAR-modified T cells on d12 after a single transduction with ROR1-CAR encoding lentivirus (MOI=3) using the tEGFR marker and found consistently higher transduction efficiencies in CD4+ compared to CD8+ CAR T cell lines obtained from the same individuals. To confirm expression of the ROR1-CAR on the surface of CD4+ T cells, we utilized biotinylated recombinant Fc-ROR1 extracellular domain fusion protein that directly binds to the scFv of the ROR1-CAR and specifically stained CD4+ T cells transduced with ROR1-CAR lentivirus but not untransduced control CD4+ T cells (FIG. 3). We enriched transgene expressing CD4+ T cells using the tEGFR marker and expanded the CAR-positive T cell subset by stimulation with anti-CD3 mAb. More than 3-log expansion of CD4+ CAR T cells could be achieved at the end of a 14-day stimulation cycle, which is equivalent to the amplification observed in CD8+ CAR CTL. After expansion, we confirmed stable expression of the ROR1-CAR on the cell surface of CD4+ CAR T cells (data not shown) and analyzed recognition of ROR1-positive tumor cells.
CD4+ ROR1-CAR T Cells Specifically Recognize ROR1-Positive Tumors
We analyzed the effector function of CD4+ ROR1-CAR T cells against ROR1-positive primary tumor cells and tumor cell lines. We analyzed the ability of CD4+ CAR T cells to confer direct cytotoxicity by chromium release assay (CRA) and detected weak but specific lysis of ROR1-positive target cells at the end of the standard 4-hour incubation (FIG. 4). We extended the CRA to 10 hours and observed a further increase in specific lysis, however, the overall cytolytic activity of CD4+ CAR T cells was still lower than CD8+ ROR1-CAR CTL (FIGS. 2 and 4). CD4+ ROR1-CAR T cells from both healthy donors and CLL patients specifically recognized primary CLL cells, the ROR1-positive tumor cell lines Jeko-1 (MCL) and BALL-1 (B-ALL), and K562 cells that were stably transfected with the ROR1-gene (K562/ROR1) but not native ROR1-negative K562 cells by IFN-γ ELISA, demonstrating specific recognition of ROR1 on the cell surface of target cells (FIG. 5A). Multiplex cytokine analysis revealed production of other Th1 cytokines such as TNF-α and IL-2 at significantly higher levels compared to CD8+ CAR CTL, and production of IL-4, IL-10 and IL-17 (FIG. 5B).
Next, we evaluated the proliferation of CD4+ CAR T cells after stimulation with ROR1-positive tumor cells by CFSE staining and used stringent culture conditions without addition of exogenous cytokines to remove any potential unspecific stimulus. CD4+ CAR T cells showed dramatic and specific proliferation in response to ROR1-positive tumor cells. Both the percentage of T cells that was induced to proliferate and the number of cell divisions that the proliferating subset performed was significantly higher in CD4+ compared to CD8+ CAR T cells (FIG. 6). Collectively, our data demonstrate that CD4+ T cells obtained from both healthy donors and CLL patients acquire anti-tumor reactivity after genetic modification with a ROR1-specific CAR. Moreover, the ability to proliferate in the absence of exogenous cytokines and to produce high levels of Th1 cytokines suggest that CD4+ CAR T cells exert typical helper functions after stimulation through the CAR and in addition to conferring direct anti-tumor effects, could also be utilized to augment CD8+ CAR CTL.
CAR-Modified, but not Untransduced CD4+ T Cells Provide Help to CD8+ CAR CTL
To analyze whether CD4+ CAR T cells are able to provide help to CD8+ CAR CTL, we performed co-culture experiments with CAR-transduced and control untransduced polyclonal CD4+ and CD8+ T cell lines that we established from healthy donors and CLL patients. As readout for provision of help, we defined an improvement in tumor-specific CD8+ effector function in the presence of CD4+ T cells compared to CD8+ T cells cultured alone. We combined either CAR-transduced or untransduced control CD4+ T cells with CD8+ CAR CTL at distinct CD4:CD8 ratios (2:1, 1:1, 1:2), stimulated them with ROR1-positive tumor cells and measured proliferation by CFSE dye dilution. We found, that the addition of CAR-transduced, but not untransduced CD4+ T cells to CD8+ CAR CTL significantly increased specific proliferation of the CD8+ subset compared to CD8+ CAR CTL alone (FIG. 7). The increase in proliferation was most pronounced, when at least an equivalent amount of CD4+ CART cells (CD4:CD8 ratio of 2:1 or 1:1) was added to the co-culture. The combination of untransduced CD4+ with untransduced CD8+ T cells served as additional control and did not induce unspecific proliferation in the CD8+ subset (data not shown).
Discussion
Gene expression profiling studies have identified genes that are preferentially or exclusively expressed by malignant but not by normal B cells and ROR1 emerged as a CLL signature gene in 2 independent analyses (27,28). Our studies illustrate the potential to target ROR1-positive malignant cells with engineered T cells expressing a ROR1-CAR. CD8 and CD4+ ROR1-CAR T cells could be derived from normal donors after lentiviral transduction of either bulk PBMCs or sort-purified T cells. CD8+ ROR1-CAR transduced T cells efficiently lysed primary B-CLL, but not normal resting or activated B-cells. CD4+ ROR1-CAR transduced T cells weakly lysed primary B-CLL, but not normal resting or activated B-cells. These T cells produced effector cytokines including TNF-α, IFNγ, IL-2, IL-4, and IL-10. CAR-transduced CD4+ T cells produced significantly higher amounts of cytokines than the transduced CD8+ cells. Both cell types were capable of proliferating in response to ROR1-expressing tumor cells. Again, CD4+ ROR1-CAR T cells proliferated 2-3 fold higher than CD8+ ROR1-CAR CTLs. These results indicate that the transduced CD4+ helper T cells exert typical helper functions suggesting they could be utilized to augment CD8+ CAR CTLs.
Example 3—the Effector Function of CD4+ ROR1-CAR T Cells from Derived from Naïve, Central and Effector Memory Subsets
The effector function of CD4 T cells derived from naïve, central and effector memory subsets and then modified with the ROR1 CAR were compared.
Materials and Methods
Sort Purification of Naïve, Central, and Effector Memory CD4 Cells
CD4+ T cells were isolated from PBMC of a healthy donor using negative magnetic bead selection (Miltenyi CD4 isolation kit) that yields untouched CD4+ T cells. The CD4+ fraction was labeled with conjugated anti-CD45RA, anti-CD45RO and anti-CD62L mAb and flow sort purified using a FACS Aria flow sorter (BD Biosciences), and naïve (CD45RA+ CD45RO− CD62L+), central memory (CD45RA− CD45RO+ CD62L+) and effector memory (CD45RA− CD45RO+ CD62L−) CD4+ T cells purified based on expression of these defined markers.
CFSE Proliferation Assay
T cells were labeled with 0.2 μM carboxyfluorescein succinimidyl ester (CFSE; Invitrogen), washed, and plated with stimulator cells at a ratio of 2:1 in CTL medium containing 10 U/mL recombinant human IL-2. After a 72-hour incubation, cells were labeled with anti-CD8 or CD4 mAb and propidium iodide (PI) to exclude dead cells from analysis. Samples were analyzed by flow cytometry, and cell division of live CD8+ and CD4+ T cells assessed by CFSE dilution.
Cytokine Assays
For analyses of cytokine secretion, target and effector cells were plated in triplicate wells at an E/T ratio of 2:1, and interferon INFγ, tumor necrosis factor (TNF-α), and IL-2 were measured by multiplex cytokine immunoassay (Luminex) in supernatant removed after a 24-hour incubation.
Results
We flow sort purified CD4+ N, central (CM) and effector memory (EM) CD4+ T cells from the peripheral blood of 3 healthy donors based on expression of CD45RA, CD45RO and CD62L (FIG. 8A), and compared their effector function after modification with the ROR1-CAR. We achieved similarly high transduction efficiencies in CAR T cell lines derived from each of the three subsets. Multiparameter flow cytometry after enrichment of transgene expressing T cells showed expression of CD45RO and loss of CD45RA in the CD4+ N CAR T cell line, consistent with an activated phenotype after the lentiviral transduction. The CD4+ N, CM and EM CAR T cell lines retained differential expression of CD62L, confirming that the initial flow sort purification had been performed with high purity.
Then, we analyzed tumor recognition, cytokine secretion and proliferation of CD4+ CAR T cells derived from N, CM and EM subsets and compared them to the CAR T cell lines generated from bulk CD4+ T cells. We observed specific recognition of ROR1-positive tumor cells by IFN-γ ELISA in each of the cell lines. Multiplex cytokine analysis revealed that CD4+ CAR T cells derived from the N subset produced by far the highest levels of Th1 cytokines, especially IL-2 (FIG. 8C) and CFSE dye dilution showed they proliferated most vigorously in response to stimulation with ROR1-positive tumor cells (FIG. 8B).
Discussion
Our studies illustrate the potential to target ROR1-positive malignant cells with engineered T cells expressing a ROR1-CAR. CD8 and CD4+ ROR1-CAR T cells could be derived from both normal donors after lentiviral transduction of either bulk PBMCs and sort-purified T cells from defined naïve or memory T cell subsets. CD4+ naïve, central memory, and effector T cells produced effector cytokines including TNFα, IFNγ, IL-2, IL-4, and IL-10. CAR-transduced CD4+ cells derived from the naïve subset produced significantly higher amounts of TNFα and IL-2 than central and effector memory derived CD4+ CAR T cells after signaling through the CAR. All CD4 cell types were capable of proliferating in response to ROR1/K562, however in the CAR-transduced CD4+ cells derived from the naïve subset, the percentage of T cells that was induced to proliferate and the number of cell divisions that the proliferating subset underwent were significantly higher. Both cytokine profile and proliferative capacity indicate that naïve CD4+ ROR1-CAR T cells may be best suited to augment CD8+ ROR1-CAR CTL.
Example 4—Naive CD4+ T Cells are Better Helpers than Memory CD4+ T Cells
Naïve, central memory, and effector transduced CD4+ T cells were co-cultured with transduced CD8+ cytotoxic T lymphocytes and the proliferative response of the cells was measured in response to stimulation with K562/ROR1 cells.
Materials and Methods
Co-Culture
Naïve, central and effector memory derived ROR1-CAR transduced CD4+ T cells and ROR1-CAR transduced CD8+ cytotoxic T lymphocytes derived from naïve and central memory CD8+ T cells were labeled with CFSE, and CD4+ and CD8+ CAR T cell lines co-cultured at a 1:1 ratio. The co-cultures were then stimulated with K562/ROR1 cells and control K562 cells and cell proliferation was measured by CFSE dye dilution assay after 5 days of incubation. For flow analysis, samples were stained with conjugated anti-CD8 and anti-CD4 mAb to distinguish CD8+ and CD4+ subsets.
Results
CD4+ Naïve CAR T Cells have a Superior Ability to Augment the Effector Function of CD8+ CAR CTL
We compared the helper function of CD4+ N, CM and EM CAR T cell lines to determine whether the favorable cytokine profile and proliferative potential of CD4+N CAR T cells would also translate into the strongest helper effect for CD8+ CAR CTL. Previous work has demonstrated that there are intrinsic differences between N, CM and EM CD8+ T cells that affect their potential utility for adoptive immunotherapy. Our group has recently shown that CM but not EM derived CD8+ T cells are able to persist for extended periods after adoptive transfer which makes them a preferred subset of CD8+ T cells for immunotherapy (33,34). Other groups suggested that CD8+ N T cells may also possess favorable traits for use in T cell therapy (35,36). Thus, we generated CD8+ CAR CTLs from sort purified N and CM T cells to determine the optimal combination of CD8+ and CD4+ CAR T cell subsets. Following lentiviral transduction and enrichment of CAR-transduced CD8+ T cells using the tEGFR marker, we confirmed tumor-reactivity of the CD8+ N, and CM CAR CTLs (data not shown) and performed co-culture experiments with CD4+ CAR T cells as before. As anticipated, co-culture of CD8+ N and CM CAR CTL with CD4+ N CAR T cells resulted in significantly higher tumor-specific proliferation of the CD8+ subset compared to co-culture with CD4+ CM or EM CAR T cells, or the CD8+ CAR CTL alone (FIG. 9). Out of all combinations, maximum proliferation of the CD8+ CAR CTL in response to stimulation with ROR1-positive tumor cells was observed after co-culture of CD4+ N CAR T cells with CD8+ CM CAR CTL (FIG. 9). Collectively, our data demonstrate that there are intrinsic differences between N, CM and EM CD4+ T cells in their cytokine profile and proliferative potential, with higher production of IL-2 and superior proliferation in CD4+ N T cells. Our data suggest that sort purified N, rather than CM, EM or bulk CD4+ T cells may be best suited to augment the effector function of CD8+ CTL, and complement previous work in CD8+ T cells that CM derived CD8+ T cells possess favorable characteristics for use in adoptive immunotherapy.
Discussion
Collectively, these data demonstrate that the adoptive transfer of ROR1-CAR modified CD4+ and CD8+ T cells confers potent anti-tumor responses in an in vivo model of aggressive systemic lymphoma and provide evidence for a beneficial and synergistic effect of CD4+ CAR T cells on the anti-tumor efficacy of CD8+ CAR CTL. Our data illustrate how the analysis of cell-intrinsic qualities can inform the rational design of cell products containing both tumor-specific CD8+ and CD4+ T cells to improve outcomes of cancer immunotherapy.
Example 5—Mouse Tumor Model of Systemic Mantle Cell Lymphoma (NSG/Jeko-1-ffLuc)
We examined the effect of providing CD4 help on the anti-tumor efficacy of ROR1-CAR modified CD8+ CTL in an in vivo model of aggressive systemic mantle cell lymphoma.
Materials and Methods
Sub-lethally irradiated NOD/SCID/gamma−/− (NSG) mice were engrafted via tail vein injection with 5×105 Jeko-1 cells that had been stably transfected with firefly luciferase (Jeko-1/ffLuc) to enable assessment of tumor burden and distribution using bioluminescence imaging. We confirmed the consistent engraftment (take rate=100%) and development of rapidly progressive disseminated lymphoma in NSG mice under these conditions. Following tumor engraftment, groups of 3 mice received either CD8+ CAR CTLs (group 1), CD4+ CART cells (group 2), a combination of CD8+ and CD4+ ROR1-CAR transduced T cells (group 3), untransduced control T cells ( group 4,5,6) via tail vein injection or no treatment (group 7). The total number of transferred T cells was 10×106 in all cases. We obtained eye bleeds from the mice 2 days after adoptive transfer and confirmed the presence of ROR1-CAR transduced or untransduced T cells in the peripheral blood.
Results
On day 6 after T-cell transfer, we performed bioluminescence imaging to evaluate tumor burden. The strongest anti-tumor effect was observed in mice that received the combination of CD8+ and CD4+ ROR1-CAR T cells, with >2 log reduction in bioluminescence signal compared to the control group (FIG. 10). We also observed a strong anti-tumor effect in mice that received either CD8+ or CD4+ ROR1-CAR modified T cells, with >1 log reduction in bioluminescence signal compared to controls (FIG. 10). Importantly, the reduction in tumor burden after administration of the CD8+/CD4+ CAR T cell combination was greater than that of the CD8+ CAR CTL and CD4+ CAR T cell groups combined suggesting that CD4+ CAR T cells and CD8+ CAR CTL were working synergistically.
Discussion
Collectively, these data demonstrate that the adoptive transfer of ROR1-CAR modified CD4+ and CD8+ T cells confers potent anti-tumor responses in an in vivo model of aggressive systemic lymphoma and provide evidence for a beneficial and synergistic effect of CD4+ CAR T cells on the anti-tumor efficacy of CD8+ CAR CTL. Our data illustrate how the analysis of cell-intrinsic qualities can inform the rational design of cell products containing both tumor-specific CD8+ and CD4+ T cells to improve outcomes of cancer immunotherapy.
Example 6—CD19 CAR T Cells Exhibit the Same Synergy
We examined the effect of providing CD4 help on the anti-tumor efficacy of CD19 modified CD8+ CTL in coculture in vitro and in an in vivo model of aggressive systemic mantle cell lymphoma.
Materials and Methods
CD19 CART cells can be prepared as described in US 2008/0131415, which is hereby incorporated by reference.
Co-Culture Assay
CD19-CAR transduced CD4+ T cells and CD19-CAR transduced CD8+ cytotoxic T lymphocytes were labeled with CFSE, and co-cultured at a 2:1, 1:1 and 1:2 ratio. The co-cultures were then stimulated with K562/ROR1 cells and control K562 cells and cell proliferation measured by CFSE dye dilution assay after 5 days of incubation. For flow analysis, samples were stained with conjugated anti-CD8 and anti-CD4 mAb to distinguish CD8+ and CD4+ subsets.
In Vivo Model
Sublethally irradiated NOD/SCID/gamma−/− (NSG) mice were engrafted via tail vein injection with 5×105 Jeko-1 cells that had been stably transfected with firefly luciferase (Jeko-1/ffLuc) to enable assessment of tumor burden and distribution using bioluminescence imaging. We confirmed the consistent engraftment (take rate=100%) and development of rapidly progressive disseminated lymphoma in NSG mice under these conditions. Following tumor engraftment, groups of 3 mice received either CD8+ CD19 CAR CTLs (group 1), CD4+ CD 19 CART cells (group 2), a combination of CD8+ and CD4+ CD19CAR transduced T cells (group 3), untransduced control T cells ( group 4,5,6) via tail vein injection or no treatment (group 7). The total number of transferred T cells was 10×106 in all cases. We obtained eye bleeds from the mice 2 days after adoptive transfer.
Results
FIG. 10 shows the superior ability of CD4+ CAR T-cell lines derived from the naïve subset to augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL in co-culture experiments with CD8+ CD19-CAR CTLs and CD4+ CD19-CAR T-cell lines, stimulated with the CD19+ mantle cell lymphoma tumor line Jeko-1. Although, CD4+ CAR T-cell lines derived from the central or effector memory subset augment tumor-specific proliferation of central memory-derived CD8+ CAR CTL to much less extent.
FIG. 11 shows that CD8+ CAR T cells and CD4+ CAR T cells independently confer direct anti-tumor efficacy in a lymphoma model in immunodeficient mice (NOD/SCID-Raji). Mice received either CD19-CAR transduced or control mock-transduced CD8+ central memory-derived (FIG. 11A), or CD19-CAR transduced or control mock-transduced CD4+ naïve-derived T cells (FIG. 11B).
FIG. 12 shows the augmentation and synergistic effect CD4+ ROR1-CAR modified T cells on the anti-tumor efficacy of CD8+ ROR1-CAR CTLs in a mouse tumor model of systemic mantle cell lymphoma (NSG/Jeko-1-ffLuc). Anti-tumor efficacy of ROR1-CAR modified CD8+ and CD4+ T cells in a mouse tumor model of systemic aggressive mantle cell lymphoma (NSG/Jeko-1) was enhanced as compared to either cell population alone or when compared to untransduced cells.
FIG. 13 shows synergy of CD8+ and CD4+ CD19-CAR T cells in a mouse model of systemic lymphoma (NSG/Raji). Engraftment of the Raji tumor was confirmed by bioluminescence imaging on day 6 after tumor inoculation (before treatment) (treatment scheme shown in FIG. 13A, tumor engraftment by bioluminescence shown in FIG. 13B). Analysis of tumor burden using bioluminescence imaging showed complete eradication of the Raji tumors in the cohorts of mice treated with CD8+ CD19-CAR T cells, and in mice treated with the combined CD8+ and CD4+ CD19-CAR T-cell product (after treatment middle black and grey bars, FIG. 13B). The mice were then challenged with a second inoculum of Raji tumor cells and the frequency of CD4+ and CD8+ CAR T cells in the peripheral blood, and tumor engraftment were analyzed. In mice treated with a combined CD8+ and CD4+ CAR T-cell product, significantly higher levels CD8+ CAR T cells after the tumor challenge (FIG. 13C, lower panels), and complete rejection of the Raji inoculum (after tumor challenge right grey bar, FIG. 13B). In contrast, in mice that had received CD8+ CD19-CAR CTL alone, we did not detect an increase in CAR T cells after the tumor challenge (FIG. 13C) and the Raji tumor cells were able to engraft (after tumor challenge right black bar, panel FIG. 13B).
Discussion
Collectively, these data demonstrate that transducing the cells with another CAR construct, CD19, CD19-CAR modified CD4+ and CD8+ T cells confer potent anti-tumor responses in an in vivo model of aggressive systemic lymphoma and provide evidence for a beneficial and synergistic effect of CD4+ CAR T cells on the anti-tumor efficacy of CD8+ CAR CTL.
The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein. All references and documents referred to herein are hereby incorporated by reference.
REFERENCES
  • 1. Cheever, M. A., et al., Specificity of adoptive chemoimmunotherapy of established syngeneic tumors. J. Immunol. 125, 711-714 (1980).
  • 2. Pahl-Seibert, M.-F. et al. Highly protective in vivo function of cytomegalovirus IE1 epitope-specific memory CD8 T cells purified by T-cell receptor-based cell sorting. J. Virol. 79, 5400-5413 (2005).
  • 3. Riddell, S R. et al. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 257, 238-241 (1992).
  • 4. Walter, E. A. et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N. Engl. J. Med. 333, 1038-1044 (1995).
  • 5. Rooney, C. M. et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 92, 1549-1555 (1998).
  • 6. Dudley, M. E. et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298, 850-854 (2002)
  • 7. Bollard, C. M. et al. Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin's disease. J. Exp. Med. 200, 1623-1633 (2004).
  • 8. Dudley, M. E. et al. Adoptive cell transfer therapy following nonmyeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J. Clin. Oncol. 23, 2346-2357 (2005).
  • 9. Gattinoni, L., Powell Jr, D. J., Rosenberg, S. A., & Restifo, N. P. Adoptive immunotherapy for cancer: building on success. Nat. Rev. Immunol. 6, 383-393 (2006).
  • 10. Blattman, J. N. & Greenberg, P. D. Cancer Immunotherapy: A treatment for the masses. Science 305, 200-205 (2004).
  • 11. Kessels, H. W. H. G. et al. Immunotherapy through TCR gene transfer. Nat. Immuno!. 2, 957-961 (2001).
  • 12. Stanislawski, T. et al. Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat. Immunol. 2, 962-970 (2001).
  • 13. Brentjens, R. J. et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279-286 (2003).
  • 14. Morgan, R. A. et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science advance online publication Aug. 31, (2006). DOI: 10.1126/science.1129003
  • 15. Bleakley, M. & Riddell, S. R. Molecules and mechanisms of the graft versus leukemia effect. Nat. Rev. Cancer 4, 371-380 (2004).
  • 16. Dudley, M. E. et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. J. Immunother. 24, 363-373 (2001).
  • 17. Yee, C. et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred cells. Proc. Natl. Acad. Sci. USA 99, 16168-16173 (2002).
  • 18. Sallusto, F. et al., Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22, 745-763 (2004).
  • 19. Butcher, E. C. & Picker, L. J. Lymphocyte homing and homeostasis. Science 272, 60-66 (1996).
  • 21. Dudley, M. E. et al. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J. Immunother. 25, 243-251 (2002).
  • 22. Gattinorti, L. et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Invest. 115, 1616-1626 (2005).
  • 23. Schmitt T M, Ciofani M, Petrie H T, Zuniga-Plucker J C. Maintenance of T cell specification and differentiation requires recurrent notch receptor-ligand interactions. J Exp Med. 2004; 200(4):469-479.
  • 24. Wang J, Press O W, Lindgren C G, et al. Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes. Mol Ther. 2004; 9(4): 577-586.
  • 25. Riddell S R, Greenberg P D. The use of anti-CD3 and anti-CD28 monoclonal antibodies to clone and expand human antigen-specific T cells. J Immunol Methods. 1990; 128(2):189-201.
  • 26. Baskar S, Kwong K Y, Hofer T, et al. Unique cell surface expression of receptor tyrosine kinase ROR1 in human B-cell chronic lymphocytic leukemia. Clin Cancer Res. 2008; 14(2):396-404.
  • 27. Klein U, Tu Y, Stolovitzky G A, et al. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 2001; 194(11):1625-1638.
  • 28. Rosenwald A, Alizadeh A A, Widhopf G, et al. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 2001;
  • 29. Fukuda T, Chen L, Endo T, et al. Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a. Proc Natl Acad Sci USA. 2008; 105(8):3047-3052.
  • 30. Lapalombella R, Andritsos L, Liu Q, et al. Lenalidomide treatment promotes CD154 expression on CLL cells and enhances production of antibodies by normal B cells through a PI3-kinase-dependent pathway. Blood. 2010; 115(13):2619-2629.
  • 31. Berger C, Jensen M C, Lansdorp P M, Gough M, Elliott C, Riddell S R. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008; 118(1):294-305.

Claims (11)

What is claimed is:
1. An adoptive cellular immunotherapy composition comprising chimeric antigen receptor-modified CD4+ T lymphocytes and chimeric antigen receptor-modified CD8+ T lymphocytes, wherein:
(a) the chimeric antigen receptor-modified CD4+ T lymphocytes in the composition consist of helper T lymphocytes that contain a chimeric antigen receptor that specifically binds to an antigen, wherein the CD4+ helper T lymphocytes are derived from: (i) a CD45RA+ CD62L+ naïve T cell enriched CD4+ population; or (ii) a CD45RA+ CD62L+ naïve T cell enriched and CD45RO+ CD62L+ central memory T cell enriched CD4+ population;
(b) the chimeric antigen receptor-modified CD8+ T lymphocytes in the composition consist of CD8+ cytotoxic T lymphocytes that are derived from a central memory-enriched CD8+ cell population and contain a chimeric antigen receptor that specifically binds to the antigen; and
wherein (a) at least 60% of the chimeric antigen receptor-modified CD4+ T lymphocytes are surface positive for CD62L and CD45RA or CD45RO; or (b) at least 80% of the chimeric antigen receptor-modified CD4+ T lymphocytes are surface positive for CD62L and CD45RA or CD45RO.
2. The adoptive cellular immunotherapy composition according to claim 1, wherein the antigen is associated with a disease or disorder selected from a solid tumor, a hematologic malignancy, a melanoma, and an infection with a pathogen.
3. The adoptive cellular immunotherapy composition according to claim 1, wherein the antigen is selected from ROR1, tEGFR, Her2, L1-CAM, CD19, CD20, CD22, mesothelin, and CEA.
4. The adoptive cellular immunotherapy composition according to claim 1, wherein the antigen is a pathogen specific cell surface antigen selected from an HIV antigen, an HCV antigen, an HBV antigen, a hepatitis B surface antigen, a CMV antigen, and a parasitic antigen.
5. The adoptive cellular immunotherapy composition according to claim 1, wherein the chimeric antigen receptor of (a) and/or (b) comprises an extracellular antibody variable domain or single-chain antibody fragment specific for an antigen associated with a disease or disorder, and an intracellular signaling module.
6. The adoptive cellular immunotherapy composition according to claim 5, wherein each of the intracellular signaling module of the chimeric antigen receptor contained by the CD4+ T lymphocytes and the intracellular signaling module of the chimeric antigen receptor contained by the CD8+ T lymphocytes, individually, comprise (a) a CD28 costimulatory domain and a CD3 intracellular signaling domain, or (b) a 4-1BB costimulatory domain and a CD3 intracellular signaling domain.
7. The adoptive cellular immunotherapy composition according to claim 1, wherein (a) the intracellular signaling domain of the chimeric antigen receptor contained by the CD8+ T lymphocytes is the same as the intracellular signaling domain of the chimeric antigen receptor contained by the CD4+ T lymphocytes, or (b) the chimeric antigen receptor contained by the CD8+ T lymphocytes is the same as the chimeric antigen receptor contained by the CD4+ T lymphocytes.
8. The adoptive cellular immunotherapy composition according to claim 1, wherein (a) the intracellular signaling domain of the chimeric antigen receptor in the CD8+ T lymphocytes is different from the intracellular signaling domain of the chimeric antigen receptor in the CD4+ T lymphocytes; or (b) the chimeric antigen receptor contained by the CD8+ T lymphocytes is different than the chimeric antigen receptor contained by the CD4+ T lymphocytes.
9. The adoptive cellular immunotherapy composition according to claim 1, wherein the CD4+ helper T lymphocytes are derived from a CD45RA+ CD62L+ naïve T cell-enriched CD4+ population.
10. The adoptive cellular immunotherapy composition according to claim 1, wherein the CD4+ helper T lymphocytes are derived from a CD45RA+ CD62L+ naïve T cell-enriched and CD45RO+ CD62L+ central memory T cell-enriched CD4+ population.
11. The adoptive cellular immunotherapy composition according to claim 9, wherein the CD4+ helper T lymphocytes augment the effector function of the CD8+ T lymphocytes.
US15/969,438 2011-03-23 2018-05-02 Method and compositions for cellular immunotherapy Active US11065278B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/969,438 US11065278B2 (en) 2011-03-23 2018-05-02 Method and compositions for cellular immunotherapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161466552P 2011-03-23 2011-03-23
PCT/US2012/030388 WO2012129514A1 (en) 2011-03-23 2012-03-23 Method and compositions for cellular immunotherapy
US201414006641A 2014-05-05 2014-05-05
US15/969,438 US11065278B2 (en) 2011-03-23 2018-05-02 Method and compositions for cellular immunotherapy

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2012/030388 Continuation WO2012129514A1 (en) 2011-03-23 2012-03-23 Method and compositions for cellular immunotherapy
US14/006,641 Continuation US9987308B2 (en) 2011-03-23 2012-03-23 Method and compositions for cellular immunotherapy

Publications (2)

Publication Number Publication Date
US20180296602A1 US20180296602A1 (en) 2018-10-18
US11065278B2 true US11065278B2 (en) 2021-07-20

Family

ID=46208746

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/006,641 Active 2034-01-24 US9987308B2 (en) 2011-03-23 2012-03-23 Method and compositions for cellular immunotherapy
US15/969,438 Active US11065278B2 (en) 2011-03-23 2018-05-02 Method and compositions for cellular immunotherapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/006,641 Active 2034-01-24 US9987308B2 (en) 2011-03-23 2012-03-23 Method and compositions for cellular immunotherapy

Country Status (16)

Country Link
US (2) US9987308B2 (en)
EP (1) EP2689010B1 (en)
JP (5) JP6203705B2 (en)
KR (1) KR101976882B1 (en)
CN (3) CN103502438A (en)
AU (3) AU2012230780B2 (en)
BR (1) BR112013024395B1 (en)
CA (1) CA2830953C (en)
ES (1) ES2841983T3 (en)
IL (1) IL228603B (en)
MX (2) MX359513B (en)
NZ (1) NZ743310A (en)
RU (1) RU2688185C2 (en)
SG (2) SG10201602253SA (en)
WO (1) WO2012129514A1 (en)
ZA (1) ZA201901167B (en)

Families Citing this family (354)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2365823B1 (en) 2008-10-30 2016-11-30 Yeda Research And Development Company Ltd. Anti third party central memory t cells, methods of producing same and use of same in transplantation and disease treatment
NZ743310A (en) * 2011-03-23 2022-11-25 Fred Hutchinson Cancer Center Method and compositions for cellular immunotherapy
MX351226B (en) 2011-09-08 2017-10-05 Yeda Res & Dev Anti third party central memory t cells, methods of producing same and use of same in transplantation and disease treatment.
DK2855667T3 (en) 2012-05-25 2023-10-30 Cellectis Methods for manipulation of allogeneic and immunosuppressant-resistant T cells for immunotherapy
JP6574381B2 (en) * 2012-08-20 2019-09-11 フレッド ハッチンソン キャンサー リサーチ センター Methods and compositions for cellular immunotherapy
US10241113B2 (en) 2012-10-10 2019-03-26 H. Lee Moffitt Cancer Center And Research Institute, Inc. CD28 expression during lenalidomide immune modulation
AU2013204922B2 (en) 2012-12-20 2015-05-14 Celgene Corporation Chimeric antigen receptors
TW201446794A (en) 2013-02-20 2014-12-16 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
ES2760023T3 (en) 2013-02-20 2020-05-12 Univ Pennsylvania Cancer treatment using humanized anti-EGFRvIII chimeric antigen receptor
CN105874061B (en) * 2013-02-26 2021-08-10 纪念斯隆-凯特琳癌症中心 Compositions and methods for immunotherapy
US10238690B2 (en) 2013-03-15 2019-03-26 Celgene Corporation Modified T lymphocytes comprising an inducible caspase and methods of apoptosis
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
BR112015026122A8 (en) 2013-04-18 2020-01-21 Armo Biosciences Inc polyethylene glycol-yl-10 agent (peg-il-10), its use, pharmaceutical composition, sterile container and kit
US11077144B2 (en) 2013-05-13 2021-08-03 Cellectis CD19 specific chimeric antigen receptor and uses thereof
LT3546572T (en) * 2013-05-13 2024-05-27 Cellectis Cd19 specific chimeric antigen receptor and uses thereof
US9823255B2 (en) 2013-06-17 2017-11-21 Armo Biosciences, Inc. Method for assessing protein identity and stability
JP6516740B2 (en) 2013-08-02 2019-05-22 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Modification of antitumor T cell immunity via stem cell and chimeric antigen receptor
AU2014311432A1 (en) 2013-08-30 2016-03-03 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
BR112016010166A2 (en) 2013-11-11 2017-12-05 Armo Biosciences Inc Methods for Using Interleukin-10 to Treat Diseases and Disorders
CN104745596B (en) * 2013-11-15 2020-02-11 中国医学科学院基础医学研究所 Cell preparation targeting hepatoma cells
MX2016008076A (en) 2013-12-19 2016-08-12 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof.
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
EP3097117B1 (en) 2014-01-21 2023-10-04 Novartis Ag Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
US10532088B2 (en) 2014-02-27 2020-01-14 Lycera Corporation Adoptive cellular therapy using an agonist of retinoic acid receptor-related orphan receptor gamma and related therapeutic methods
CN111514283A (en) 2014-04-07 2020-08-11 诺华股份有限公司 Treatment of cancer using anti-CD 19 chimeric antigen receptors
KR20230038310A (en) 2014-04-10 2023-03-17 시애틀 칠드런즈 호스피탈 디/비/에이 시애틀 칠드런즈 리서치 인스티튜트 Method and compositions for cellular immunotherapy
CN111961647A (en) 2014-04-16 2020-11-20 朱诺治疗有限公司 Methods, kits and devices for expanding cell populations
ES2759260T3 (en) * 2014-04-23 2020-05-08 Juno Therapeutics Inc Methods for Isolating, Cultivating, and Genetically Modifying Immune Cell Populations for Adoptive Therapy
EP3998278A1 (en) 2014-04-25 2022-05-18 2seventy bio, Inc. Mnd promoter chimeric antigen receptors
JP6523337B2 (en) 2014-05-05 2019-05-29 リセラ・コーポレイションLycera Corporation Benzenesulfonamides and related compounds for use as agonists of ROR.gamma. And disease treatment
WO2015171610A2 (en) 2014-05-05 2015-11-12 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of rory and the treatment of disease
WO2015187295A2 (en) 2014-06-02 2015-12-10 Armo Biosciences, Inc. Methods of lowering serum cholesterol
SG10202108458XA (en) 2014-06-06 2021-09-29 2Seventy Bio Inc Improved t cell compositions
WO2016011210A2 (en) * 2014-07-15 2016-01-21 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
SG11201700418VA (en) 2014-07-21 2017-02-27 Novartis Ag Treatment of cancer using a cll-1 chimeric antigen receptor
BR112017001242A2 (en) 2014-07-21 2017-12-05 Novartis Ag cancer treatment using a cd33 chimeric antigen receptor
MX2017001011A (en) 2014-07-21 2018-05-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor.
JP2017528433A (en) * 2014-07-21 2017-09-28 ノバルティス アーゲー Low immunoenhancing dose of mTOR inhibitor and CAR combination
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
EP3172231B1 (en) 2014-07-24 2021-05-05 Bluebird Bio, Inc. Bcma chimeric antigen receptors
MX367787B (en) 2014-07-29 2019-09-06 Cellectis Ror1(ntrkr1)specific chimeric antigen receptors for cancer immunotherapy.
WO2016016343A1 (en) 2014-07-31 2016-02-04 Cellectis Ror1 specific multi-chain chimeric antigen receptor
BR112017003104A2 (en) 2014-08-19 2017-12-05 Novartis Ag cancer treatment using an anti-cd123 chimeric antigen receptor
TWI805109B (en) 2014-08-28 2023-06-11 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
ES2777305T3 (en) 2014-09-04 2020-08-04 Cellectis Trophoblastic glycoprotein-specific chimeric antigen receptors (5T4, TPBG) for cancer immunotherapy
CN114621969A (en) 2014-09-17 2022-06-14 诺华股份有限公司 Targeted cytotoxic cells with chimeric receptors for adoptive immunotherapy
JP6657195B2 (en) 2014-09-19 2020-03-04 シティ・オブ・ホープCity of Hope Co-stimulatory chimeric antigen receptor T cells targeting L13Rα2
CN106973568B (en) * 2014-10-08 2021-07-23 诺华股份有限公司 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
ES2879612T3 (en) 2014-10-20 2021-11-22 Juno Therapeutics Inc Methods and compositions for dosage in adoptive cell therapy
CN107106655A (en) 2014-10-22 2017-08-29 阿尔莫生物科技股份有限公司 The method that disease and illness are treated using interleukin 10
CN107106611B (en) * 2014-10-27 2021-11-23 弗雷德哈钦森癌症研究中心 Compositions and methods for improving the efficacy of adoptive cellular immunotherapy
CA2966538A1 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
AU2015358400B2 (en) 2014-12-03 2020-09-10 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
LT3230321T (en) 2014-12-12 2019-12-10 Bluebird Bio Inc Bcma chimeric antigen receptors
EP3240803B1 (en) 2014-12-29 2021-11-24 Novartis AG Methods of making chimeric antigen receptor-expressing cells
CN105802975B (en) * 2014-12-31 2020-04-14 浙江大学 Cell preparation targeting HER2 positive tumor and application thereof
CN105802909B (en) * 2014-12-31 2021-01-01 中国医学科学院基础医学研究所 T cell preparation with HER2 specific TCR and uses thereof
MA41346A (en) 2015-01-12 2017-11-21 Juno Therapeutics Inc POST-TRANSCRIPTIONAL REGULATORY ELEMENTS OF MODIFIED HEPATITIS
EP3760644A1 (en) 2015-01-16 2021-01-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
WO2016115482A1 (en) 2015-01-16 2016-07-21 Novartis Pharma Ag Phosphoglycerate kinase 1 (pgk) promoters and methods of use for expressing chimeric antigen receptor
DK3250606T3 (en) * 2015-01-26 2021-01-04 Cellectis ANTI-CLL1-specific single-chain chimeric antigen receptors (SCCARS) for cancer immunotherapy
CA2973884A1 (en) 2015-01-30 2016-08-04 The Regents Of The University Of California Protein delivery in primary hematopoietic cells
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
CN107531805A (en) * 2015-03-05 2018-01-02 弗雷德哈钦森癌症研究中心 Immunological regulation fusion protein and application thereof
CN114958764A (en) 2015-04-08 2022-08-30 诺华股份有限公司 CD20 therapy, CD22 therapy, and combination therapy with CD19 Chimeric Antigen Receptor (CAR) -expressing cells
MX2017013247A (en) * 2015-04-15 2018-08-15 Prospect Chartercare Rwmc Llc D/B/A Roger Williams Medical Center Hepatic arterial infusion of car-t cells.
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
JP7114457B2 (en) 2015-04-17 2022-08-08 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods for Improving Efficacy and Growth of Chimeric Antigen Receptor-Expressing Cells
WO2016179343A1 (en) 2015-05-05 2016-11-10 Lycera Corporation DIHYDRO-2H-BENZO[b][1,4]OXAZINE SULFONAMIDE AND RELATED COMPOUNDS FOR USE AS AGONISTS OF RORy AND THE TREATMENT OF DISEASE
AU2016263513A1 (en) 2015-05-20 2017-11-23 Cellectis Anti-GD3 specific chimeric antigen receptors for cancer immunotherapy
KR20180020141A (en) 2015-05-28 2018-02-27 아르모 바이오사이언시스 인코포레이티드 PEGylated interleukin-10 for cancer treatment
CA2986060A1 (en) 2015-05-29 2016-12-08 Valerie Odegard Composition and methods for regulating inhibitory interactions in genetically engineered cells
MX2017016134A (en) 2015-06-11 2018-08-15 Lycera Corp Aryl dihydro-2h-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of rory and the treatment of disease.
MA42895A (en) 2015-07-15 2018-05-23 Juno Therapeutics Inc MODIFIED CELLS FOR ADOPTIVE CELL THERAPY
CN114457039A (en) * 2015-07-16 2022-05-10 耶达研究及发展有限公司 Genetically modified anti-third party central memory T cells and their use in immunotherapy
GB201512733D0 (en) * 2015-07-20 2015-08-26 Genagon Therapeutics Ab Therapeutic agents for treating conditions associated with elevated GDF15
US20190175648A1 (en) * 2015-07-21 2019-06-13 City Of Hope T cells for expression of chimeric antigen receptors and other receptors
CA2992551A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
JP7409773B2 (en) 2015-07-31 2024-01-09 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Modified cells and methods of treatment
GB201513540D0 (en) 2015-07-31 2015-09-16 King S College London Therapeutic agents
WO2017027291A1 (en) 2015-08-07 2017-02-16 Seattle Children's Hospital (dba Seattle Children's Research Institute) Bispecific car t-cells for solid tumor targeting
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
WO2017035232A1 (en) 2015-08-25 2017-03-02 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
JP6905163B2 (en) 2015-09-03 2021-07-21 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Biomarkers that predict cytokine release syndrome
EP3347026A4 (en) * 2015-09-09 2019-05-08 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Genetic engineering of macrophages for immunotherapy
US11142565B2 (en) * 2015-09-24 2021-10-12 Abvitro Llc Broadly neutralizing anti-HIV-1 antibodies that bind to an N-glycan epitope on the envelope
MX2018003534A (en) 2015-09-25 2019-04-25 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences.
MA45489A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, ASSOCIATED KITS AND APPARATUS
MA45488A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, KITS AND APPARATUS
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
ES2909973T3 (en) * 2015-11-04 2022-05-10 Hope City Chimeric receptors for the antigen that target HER2
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
MA44314A (en) 2015-11-05 2018-09-12 Juno Therapeutics Inc CHEMERICAL RECEPTORS CONTAINING TRAF-INDUCING DOMAINS, AND ASSOCIATED COMPOSITIONS AND METHODS
EP4212547A1 (en) 2015-12-03 2023-07-19 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
MA43378A (en) 2015-12-03 2018-10-10 Juno Therapeutics Inc COMPOSITIONS AND METHODS FOR REDUCTION OF IMMUNE RESPONSE AGAINST CHEMERIC ANTIGEN RECEPTORS
EP3384294B1 (en) 2015-12-04 2021-10-13 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
EP3393504A1 (en) 2015-12-22 2018-10-31 Novartis AG Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
GB201600328D0 (en) 2016-01-08 2016-02-24 Univ Oslo Hf Anti-CD37 chimeric antigen receptors and immune cells expressing them
CN108697776A (en) 2016-01-11 2018-10-23 阿尔莫生物科技股份有限公司 Interleukin-10 in generating antigentic specificity CD8+T cells and its application method
CN106978442B (en) * 2016-01-18 2020-06-19 爱康得生物医学技术(苏州)有限公司 Preparation method of chimeric antigen receptor T cell
CN108699149B (en) 2016-02-02 2023-01-06 弗雷德哈钦森癌症中心 anti-ROR 1 antibodies and uses thereof
EA201891717A1 (en) 2016-02-23 2019-02-28 Иммьюн Дизайн Корп. PREPARATIONS OF MULTI-GENOMIC RETROVIRAL VECTORS AND METHODS AND SYSTEMS FOR THEIR RECEPTION AND APPLICATION
MA43759A (en) 2016-03-16 2018-11-28 Jason Connor PROCESSES FOR THE ADAPTIVE DESIGN OF A TREATMENT REGIME AND ASSOCIATED TREATMENTS
WO2017161208A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
MX2018010415A (en) 2016-03-18 2018-11-29 Hutchinson Fred Cancer Res Compositions and methods for cd20 immunotherapy.
US11518814B2 (en) 2016-03-22 2022-12-06 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
EP3432924A1 (en) 2016-03-23 2019-01-30 Novartis AG Cell secreted minibodies and uses thereof
ES2930058T3 (en) 2016-04-01 2022-12-05 Kite Pharma Inc Chimeric receptors and methods of using the same
BR112018070073A2 (en) 2016-04-01 2019-02-12 Kite Pharma, Inc. chimeric antigen and t-cell receptors and methods of use
JP2019516352A (en) 2016-04-01 2019-06-20 アムジエン・インコーポレーテツド Chimeric receptor for FLT3 and method of using the same
EP3436036A4 (en) 2016-04-01 2020-03-18 Kite Pharma, Inc. Bcma binding molecules and methods of use thereof
CN108884459B (en) * 2016-04-26 2024-04-02 科济生物医药(上海)有限公司 Method for improving immune response cell function
WO2017192924A1 (en) 2016-05-04 2017-11-09 Fred Hutchinson Cancer Research Center Cell-based neoantigen vaccines and uses thereof
MX2018013445A (en) 2016-05-06 2019-09-09 Juno Therapeutics Inc Genetically engineered cells and methods of making the same.
MX2018013963A (en) * 2016-05-25 2019-08-22 Council Queensland Inst Medical Res Methods of immunotherapy.
CA3025523A1 (en) 2016-05-27 2017-11-30 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
US20210177896A1 (en) 2016-06-02 2021-06-17 Novartis Ag Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2017210689A1 (en) 2016-06-03 2017-12-07 Memorial Sloan-Kettering Cancer Center Adoptive cell therapies as early treatment options
MA45341A (en) 2016-06-06 2019-04-10 Hutchinson Fred Cancer Res METHODS FOR TREATING B-LYMPHOCYTE MALIGNITIES USING ADOPTIVE CELL THERAPY
KR20190039085A (en) * 2016-06-17 2019-04-10 마젠타 테라퓨틱스 인코포레이티드 Compositions and methods for depletion of cells
MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
KR20230107408A (en) 2016-07-29 2023-07-14 주노 쎄러퓨티크스 인코퍼레이티드 Anti-idiotypic antibodies against anti-cd19 antibodies
ES2951552T3 (en) 2016-07-29 2023-10-23 Juno Therapeutics Inc Immunomodulatory polypeptides and related compositions and methods
AU2017301881A1 (en) 2016-07-29 2019-02-07 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
CN106279432B (en) 2016-08-10 2019-09-20 深圳市再生之城生物医药技术有限公司 A kind of VC-CAR molecule and the application in removing HIV-1 infection cell
RU2755725C2 (en) 2016-09-12 2021-09-20 Джуно Терапьютикс, Инк. Assembly units of perfusion bioreactor bags
WO2018052828A1 (en) 2016-09-14 2018-03-22 Janssen Biotech, Inc. Chimeric antigen receptors comprising bcma-specific fibronectin type iii domains and uses thereof
CN110087657A (en) 2016-09-28 2019-08-02 阿托莎遗传股份有限公司 The method of adoptive cellular treatment
US11072660B2 (en) 2016-10-03 2021-07-27 Juno Therapeutics, Inc. HPV-specific binding molecules
MX2019003886A (en) 2016-10-07 2019-08-05 Novartis Ag Chimeric antigen receptors for the treatment of cancer.
US11896615B2 (en) 2016-10-13 2024-02-13 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
GB2605883B (en) 2016-10-18 2023-03-15 Univ Minnesota Tumor infiltrating lymphocytes and methods of therapy
JP7100028B2 (en) 2016-10-20 2022-07-12 セルジーン コーポレイション Cereblon-based heterodimerizable chimeric antigen receptor
CA3040914A1 (en) 2016-11-03 2018-05-24 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
CA3045339A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
BR112019011025A2 (en) 2016-12-03 2019-10-08 Juno Therapeutics Inc T cell modulation methods
ES2961666T3 (en) 2016-12-03 2024-03-13 Juno Therapeutics Inc Methods to determine CAR-T cell dosage
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
WO2018111763A1 (en) 2016-12-12 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Chimeric transcription factor variants with augmented sensitivity to drug ligand induction of transgene expression in mammalian cells
EP3568414A1 (en) 2017-01-10 2019-11-20 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
GB201700567D0 (en) 2017-01-12 2017-03-01 Genagon Therapeutics Ab Therapeutic agents
GB201700553D0 (en) 2017-01-12 2017-03-01 Genagon Therapeutics Ab Therapeutic agents
CN110392736A (en) 2017-01-18 2019-10-29 耶达研究及发展有限公司 The veto cell of genetic modification and its purposes in immunization therapy
US10751368B2 (en) 2017-01-18 2020-08-25 Yeda Research And Development Co. Ltd. Methods of transplantation and disease treatment
CN110461876B (en) 2017-01-20 2024-05-17 海德堡医药研究有限责任公司 Compositions and methods for depleting cd137+ cells
MX2019008538A (en) 2017-01-20 2019-11-05 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods.
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
CN116712458A (en) * 2017-02-07 2023-09-08 学校法人埼玉医科大学 Immunological biomarkers for predicting clinical outcome of cancer immunotherapy
WO2018148180A2 (en) 2017-02-07 2018-08-16 Immune Design Corp. Materials and methods for identifying and treating cancer patients
EP4008728A1 (en) 2017-02-14 2022-06-08 Kite Pharma, Inc. Cd70 binding molecules and methods of use thereof
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
BR112019017767A2 (en) 2017-02-27 2020-04-07 Juno Therapeutics Inc compositions, articles of manufacture and methods related to dosing in cell therapy
JP7178355B2 (en) 2017-02-28 2022-11-25 エンドサイト・インコーポレイテッド Compositions and methods for CAR T cell therapy
US20180280437A1 (en) 2017-03-13 2018-10-04 Kite Pharma, Inc. Chimeric antigen receptors for melanoma and uses thereof
SG11201908271WA (en) 2017-03-14 2019-10-30 Juno Therapeutics Inc Methods for cryogenic storage
JP2020511136A (en) 2017-03-17 2020-04-16 フレッド ハッチンソン キャンサー リサーチ センター Immunomodulatory fusion proteins and uses thereof
RU2019133280A (en) 2017-03-22 2021-04-22 Новартис Аг COMPOSITIONS AND METHODS FOR IMMUNO ONCOLOGY
CN107082811B (en) * 2017-03-28 2021-01-15 中山大学 Composite protein of chimeric antigen receptor and fusion induced apoptosis enzyme
AR111360A1 (en) 2017-04-03 2019-07-03 Kite Pharma Inc TREATMENT USING CHEMERIC RECEIVER T-CELLS INCORPORATING OPTIMIZED POLYFUNCTIONAL T-CELLS
CA3056261A1 (en) 2017-04-07 2018-10-11 Juno Therapeutics, Inc. Engineered cells expressing prostate-specific membrane antigen (psma) or a modified form thereof and related methods
MX2019012189A (en) 2017-04-14 2020-12-10 Juno Therapeutics Inc Methods for assessing cell surface glycosylation.
ES2912383T3 (en) 2017-04-18 2022-05-25 Fujifilm Cellular Dynamics Inc Antigen-specific immune effector cells
WO2018195339A1 (en) 2017-04-19 2018-10-25 Board Of Regents, The University Of Texas System Immune cells expressing engineered antigen receptors
JOP20180042A1 (en) 2017-04-24 2019-01-30 Kite Pharma Inc Humanized Antigen-Binding Domains and Methods of Use
KR20230164219A (en) 2017-04-27 2023-12-01 주노 테라퓨틱스 게엠베하 Oligomeric particle reagents and methods of use thereof
PT3618842T (en) 2017-05-01 2024-01-12 Juno Therapeutics Inc Combination of a cell therapy and an immunomodulatory compound
CA3064018A1 (en) 2017-05-26 2018-11-29 Kite Pharma, Inc. Methods of making and using embryonic mesenchymal progenitor cells
KR20200054160A (en) 2017-06-02 2020-05-19 주노 쎄러퓨티크스 인코퍼레이티드 Preparation and method of articles for treatment with adoptive cell therapy
AU2018275891A1 (en) 2017-06-02 2019-12-12 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
CN110997902B (en) 2017-06-20 2021-11-30 居里研究所 SUV39H1 deficient immune cells
EP3641789A4 (en) 2017-06-22 2021-08-11 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
KR20200022467A (en) 2017-06-28 2020-03-03 리제너론 파아마슈티컬스, 인크. Anti-human papilloma virus (HPV) antigen-binding protein and methods of use thereof
CA3067602A1 (en) 2017-06-29 2019-01-03 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
WO2019006418A2 (en) 2017-06-30 2019-01-03 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
MA49652A (en) 2017-07-17 2020-05-27 Janssen Biotech Inc ANTIGEN-BINDING REGIONS DIRECTED TO FIELDS OF FIBRONECTIN TYPE III AND THEIR METHODS OF USE
MX2020000900A (en) 2017-07-29 2021-01-08 Juno Therapeutics Inc Reagents for expanding cells expressing recombinant receptors.
WO2019028051A1 (en) 2017-07-31 2019-02-07 Lentigen Technology, Inc. Compositions and methods for treating cancer with anti-cd19/cd20 immunotherapy
MX2020001491A (en) 2017-08-09 2020-08-06 Juno Therapeutics Inc Methods and compositions for preparing genetically engineered cells.
US11851678B2 (en) * 2017-08-09 2023-12-26 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019046832A1 (en) 2017-09-01 2019-03-07 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
US20200292526A1 (en) 2017-09-07 2020-09-17 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
KR20200055037A (en) 2017-09-19 2020-05-20 메사추세츠 인스티튜트 오브 테크놀로지 Compositions and uses for chimeric antigen receptor T cell therapy
CN109517820B (en) 2017-09-20 2021-09-24 北京宇繁生物科技有限公司 gRNA of target HPK1 and HPK1 gene editing method
US11952408B2 (en) 2017-10-03 2024-04-09 Juno Therapeutics, Inc. HPV-specific binding molecules
CN111479613A (en) 2017-10-18 2020-07-31 凯德药业股份有限公司 Methods of administering chimeric antigen receptor immunotherapy
US20210198359A1 (en) 2017-10-25 2021-07-01 Actinium Pharmaceuticals, Inc. Anti-cd45-based lymphodepletion methods and uses thereof in conjction with act-based cancer therapies
AU2018360801A1 (en) 2017-11-01 2020-05-14 Celgene Corporation Process for producing a T cell composition
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089884A2 (en) 2017-11-01 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
BR112020008323A2 (en) 2017-11-01 2020-11-03 Juno Therapeutics Inc antibodies and chimeric antigen receptors specific for b cell maturation antigen
BR112020008638A2 (en) 2017-11-01 2020-10-20 Juno Therapeutics Inc chimeric antigen receptors specific for b cell maturation antigens (bcma)
MX2020004240A (en) 2017-11-01 2020-09-25 Juno Therapeutics Inc Process for generating therapeutic compositions of engineered cells.
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
CN111556893A (en) 2017-11-06 2020-08-18 爱迪塔斯医药股份有限公司 Methods, compositions, and components for CRISPR-CAS9 editing of CBLB in immunotherapy T cells
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
US20200330983A1 (en) 2017-11-10 2020-10-22 Juno Therapeutics, Inc. Closed-system cryogenic vessels
EP3710471A1 (en) 2017-11-16 2020-09-23 Kite Pharma, Inc. Modified chimeric antigen receptors and methods of use
KR20200116081A (en) 2017-12-01 2020-10-08 주노 쎄러퓨티크스 인코퍼레이티드 Methods of administration and control of genetically engineered cells
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
AU2018379092A1 (en) 2017-12-08 2020-06-25 Juno Therapeutics, Inc. Process for producing a composition of engineered T cells
BR112020011223A2 (en) 2017-12-08 2020-11-17 Juno Therapeutics Inc phenotypic markers for cell therapy and related methods
JP2021506260A (en) 2017-12-15 2021-02-22 ジュノー セラピューティクス インコーポレイテッド Anti-CCT5 binding molecule and how to use it
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
MX2020007543A (en) 2018-01-15 2020-09-09 Pfizer Methods of administering chimeric antigen receptor immunotherapy in combination with 4-1bb agonist.
EP3743082A4 (en) 2018-01-22 2021-11-03 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Methods of use for car t cells
WO2019152743A1 (en) 2018-01-31 2019-08-08 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
EP3746569A1 (en) 2018-01-31 2020-12-09 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
SG11202007441TA (en) * 2018-02-06 2020-09-29 Seattle Childrens Hospital Dba Seattle Childrens Res Inst Closed-system manufacturing process for car-t cells
CN112534044A (en) 2018-02-16 2021-03-19 凯德药业股份有限公司 Modified pluripotent stem cells and methods of making and using
EP3762012A1 (en) 2018-03-09 2021-01-13 Ospedale San Raffaele S.r.l. Il-1 antagonist and toxicity induced by cell therapy
WO2019195492A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
PE20201345A1 (en) 2018-04-05 2020-11-25 Juno Therapeutics Inc RECEIVERS OF T-CELLS, AND DESIGNED CELLS THAT EXPRESS THEM
JOP20200259A1 (en) 2018-04-10 2020-10-11 Kite Pharma Inc Chimeric receptors to dll3 and methods of use thereof
SG11202009881WA (en) 2018-04-12 2020-11-27 Kite Pharma Inc Chimeric receptor t cell treatment using characteristics of the tumor microenvironment
EP3787751A1 (en) 2018-05-03 2021-03-10 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
CA3100724A1 (en) 2018-06-13 2019-12-19 Novartis Ag B-cell maturation antigen protein (bcma) chimeric antigen receptors and uses thereof
US20210155941A1 (en) 2018-06-22 2021-05-27 Kite Pharma Eu B.V. Compositions and methods for making engineered t cells
AU2019301126A1 (en) 2018-07-11 2021-02-04 Celgene Corporation Uses of anti-BCMA chimeric antigen receptors
US20210277148A1 (en) 2018-07-18 2021-09-09 Amgen Inc. Chimeric receptors to steap1 and methods of use thereof
JP7404335B2 (en) 2018-07-19 2023-12-25 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Chimeric antigen receptor with BCMA specificity and its use
WO2020021114A1 (en) * 2018-07-27 2020-01-30 Nordwest Polybiocept Bioscience Gmbh Method for the treatment of a tumor patient with adoptive t cell immunotherapy
KR20210038922A (en) 2018-08-02 2021-04-08 카이트 파마 인코포레이티드 Chimeric antigen receptor therapy T cell expansion kinetics and uses thereof
MX2021001519A (en) 2018-08-09 2021-05-27 Juno Therapeutics Inc Methods for assessing integrated nucleic acids.
CA3108657A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
CN110819589B (en) * 2018-08-13 2022-10-11 上海科技大学 Method for enhancing immune effector cell function
EP3850366A1 (en) 2018-09-11 2021-07-21 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
CN112771071A (en) 2018-09-28 2021-05-07 麻省理工学院 Collagen-localized immunomodulatory molecules and methods thereof
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
JP2022512789A (en) * 2018-10-31 2022-02-07 ベリカム ファーマシューティカルズ, インコーポレイテッド T cells with suicide switch
CA3117568A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
US20210393690A1 (en) 2018-11-01 2021-12-23 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
ES2968737T3 (en) 2018-11-06 2024-05-13 Juno Therapeutics Inc Process to produce genetically engineered T cells
KR20210111247A (en) 2018-11-08 2021-09-10 주노 쎄러퓨티크스 인코퍼레이티드 Methods and Combinations for Treatment and Modulation of T Cells
SG11202105084VA (en) 2018-11-16 2021-06-29 Juno Therapeutics Inc Methods of dosing engineered t cells for the treatment of b cell malignancies
EP3883955A1 (en) 2018-11-19 2021-09-29 Board of Regents, The University of Texas System A modular, polycistronic vector for car and tcr transduction
CN113272427A (en) 2018-11-28 2021-08-17 得克萨斯大学体系董事会 Multiple genome editing of immune cells to enhance functionality and resistance to suppressive environments
AU2019386830A1 (en) 2018-11-29 2021-06-24 Board Of Regents, The University Of Texas System Methods for ex vivo expansion of natural killer cells and use thereof
FI3886894T3 (en) 2018-11-30 2024-05-24 Juno Therapeutics Inc Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
CA3120869A1 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020109953A1 (en) 2018-11-30 2020-06-04 Janssen Biotech, Inc. Gamma delta t cells and uses thereof
BR112021011063A2 (en) 2018-12-10 2021-08-31 Amgen Inc. MUTATED PIGGYBAC TRANSPOSASE
JP7386382B2 (en) 2018-12-12 2023-11-27 カイト ファーマ インコーポレイテッド Chimeric antigen receptors and T cell receptors and methods of use
AR119683A1 (en) 2019-01-29 2022-01-05 Juno Therapeutics Inc ANTIBODIES AND CHIMERIC ANTIGEN RECEPTORS SPECIFIC TO ORPHAN RECEPTOR TYROSINE KINASE-LIKE 1 (ROR1)
MX2021010441A (en) 2019-03-01 2021-09-21 Allogene Therapeutics Inc Dll3 targeting chimeric antigen receptors and binding agents.
BR112021021178A2 (en) 2019-04-26 2022-03-15 Allogene Therapeutics Inc Rituximab-resistant chimeric antigen receptors and uses thereof
CN113748201A (en) 2019-04-26 2021-12-03 艾洛基治疗公司 Methods of making allogeneic CAR T cells
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
SG11202111360YA (en) 2019-05-01 2021-11-29 Juno Therapeutics Inc Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
KR20220005075A (en) 2019-05-03 2022-01-12 카이트 파마 인코포레이티드 Methods of Administration of Chimeric Antigen Receptor Immunotherapy
BR112021024404A2 (en) 2019-06-07 2022-04-19 Juno Therapeutics Inc Automated t cell culture
SG11202113356XA (en) 2019-06-12 2021-12-30 Juno Therapeutics Inc Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
EP3990491A1 (en) 2019-06-26 2022-05-04 Massachusetts Institute of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
CA3146895A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
BR112022001194A2 (en) 2019-07-24 2022-06-07 Regeneron Pharma Chimeric antigen receptors with mage a4 specificity and uses thereof
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
BR112022003471A2 (en) 2019-08-27 2022-05-24 Janssen Biotech Inc Chimeric antigen receptor system and uses thereof
JP2022546396A (en) 2019-08-30 2022-11-04 ジュノー セラピューティクス インコーポレイテッド Machine learning method for classifying cells
BR112022003790A2 (en) 2019-09-02 2022-05-31 Inst Nat Sante Rech Med Method for selecting a tumor neoantigenic peptide, tumor neoantigenic peptide, isolated tumor neoantigenic peptide, population of autologous dendritic cells, vaccine or immunogenic composition, antibodies, method of producing an antibody, t-cell receptor, polynucleotide, vector, immune cell, cell t, neoantigenic peptide and immune cell population
EP4025597A2 (en) * 2019-09-06 2022-07-13 Avectas Limited Engineering of immune cells for ex vivo cell therapy applications
CN114729368A (en) 2019-09-09 2022-07-08 斯克里贝治疗公司 Compositions and methods for immunotherapy
WO2021061648A1 (en) 2019-09-23 2021-04-01 Massachusetts Institute Of Technology Methods and compositions for stimulation of endogenous t cell responses
US20220401539A1 (en) 2019-10-22 2022-12-22 Institut Curie Immunotherapy Targeting Tumor Neoantigenic Peptides
BR112022008023A2 (en) 2019-10-30 2022-07-12 Juno Therapeutics Gmbh CELL SELECTION AND/OR STIMULATION DEVICES AND USE METHODS
IL292566A (en) 2019-11-05 2022-06-01 Celgene Corp Uses of anti-bcma chimeric antigen receptors
CN114980918A (en) 2019-11-07 2022-08-30 朱诺治疗学股份有限公司 Combination of T cell therapy with (S) -3- [4- (4-morpholin-4-ylmethyl-benzyloxy) -1-oxo-1, 3-dihydro-isoindol-2-yl ] -piperidine-2, 6-dione
AR120563A1 (en) 2019-11-26 2022-02-23 Novartis Ag CD19 AND CD22 CHIMERIC ANTIGEN RECEPTORS AND THEIR USES
KR20220122653A (en) 2019-12-06 2022-09-02 주노 쎄러퓨티크스 인코퍼레이티드 Anti-idiotypic Antibodies and Related Compositions and Methods Against BPCMA-Target Binding Domains
CN115335399A (en) 2019-12-06 2022-11-11 朱诺治疗学股份有限公司 Anti-idiotype antibodies directed against GPRC5D target binding domains and related compositions and methods
AU2020395318A1 (en) 2019-12-06 2022-06-09 Juno Therapeutics, Inc. Methods related to toxicity and response associated with cell therapy for treating B cell malignancies
AU2021209940A1 (en) 2020-01-24 2022-08-04 Juno Therapeutics, Inc. Methods for dosing and treatment of follicular lymphoma and marginal zone lymphoma in adoptive cell therapy
CN115427550A (en) 2020-01-28 2022-12-02 朱诺治疗学股份有限公司 T cell transduction methods
KR20220152220A (en) 2020-02-12 2022-11-15 주노 쎄러퓨티크스 인코퍼레이티드 CD19-directed chimeric antigen receptor T cell composition and methods and uses thereof
MX2022009830A (en) 2020-02-12 2022-10-28 Juno Therapeutics Inc Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof.
US20230106973A1 (en) 2020-02-17 2023-04-06 Board Of Regents, The University Of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
BR112022017419A2 (en) 2020-03-03 2022-10-18 Janssen Biotech Inc GAMMA-DELTA T CELLS AND USES THEREOF
AU2021232853A1 (en) 2020-03-10 2022-09-22 Massachusetts Institute Of Technology Compositions and methods for immunotherapy of NPM1c-positive cancer
AU2021236145A1 (en) 2020-03-10 2022-09-22 Dana-Farber Cancer Institute, Inc. Methods for generating engineered memory-like NK cells and compositions thereof
US20230149462A1 (en) 2020-04-10 2023-05-18 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
US20210338833A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Chimeric antigen receptor-targeting ligands and uses thereof
WO2021221783A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Methods for identifying chimeric antigen receptor-targeting ligands and uses thereof
US11826386B2 (en) 2020-05-05 2023-11-28 Regeneron Pharmaceuticals, Inc. Compositions and methods for treating cancer
EP4149952A1 (en) 2020-05-12 2023-03-22 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
US20230178239A1 (en) 2020-05-13 2023-06-08 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
EP4153301A2 (en) 2020-05-21 2023-03-29 Board of Regents, The University of Texas System T cell receptors with vgll1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
EP4188395A1 (en) 2020-07-30 2023-06-07 Institut Curie Immune cells defective for socs1
US20230324408A1 (en) 2020-08-05 2023-10-12 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
JP2023538012A (en) 2020-08-14 2023-09-06 カイト ファーマ インコーポレイテッド Improving immune cell function
KR20230113755A (en) 2020-11-04 2023-08-01 셀진 코포레이션 CAR T cell therapy in patients receiving prior cancer alkylating agent therapy
EP4243839A1 (en) 2020-11-13 2023-09-20 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
KR20230117383A (en) 2020-12-04 2023-08-08 셀진 코포레이션 Use of Chimeric Antigen Receptor (CAR) T-Cell Therapy in Combination with Inhibitors of Inflammation-Related Soluble Factors
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
CN116917737A (en) 2021-01-10 2023-10-20 凯德药业股份有限公司 T cell therapy
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
WO2022165133A1 (en) 2021-01-28 2022-08-04 Allogene Therapeutics, Inc. Methods for transducing immune cells
EP4284512A1 (en) 2021-01-28 2023-12-06 Regeneron Pharmaceuticals, Inc. Compositions and methods for treating cytokine release syndrome
WO2022178243A1 (en) 2021-02-20 2022-08-25 Kite Pharma, Inc. Gene markers for sellecting immunotherapies
EP4301755A1 (en) 2021-03-03 2024-01-10 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
JP2024510217A (en) 2021-03-11 2024-03-06 アンスティテュ・クリー Transmembrane neoantigen peptide
EP4305053A1 (en) 2021-03-11 2024-01-17 Kite Pharma, Inc. Improving immune cell function
KR20230172047A (en) 2021-03-11 2023-12-21 므네모 테라퓨틱스 Tumor neoantigen peptides and uses thereof
AU2022233019A1 (en) 2021-03-11 2023-09-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Tumor neoantigenic peptides
KR20230159851A (en) 2021-03-22 2023-11-22 주노 쎄러퓨티크스 인코퍼레이티드 How to Determine the Potency of a Therapeutic Cell Composition
EP4314280A1 (en) 2021-03-22 2024-02-07 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
KR20240005700A (en) 2021-03-29 2024-01-12 주노 쎄러퓨티크스 인코퍼레이티드 Dosing and Treatment Methods Using Combination of Checkpoint Inhibitor Therapy and CAR T Cell Therapy
IL307257A (en) 2021-03-29 2023-11-01 Juno Therapeutics Inc Combination of a car t cell therapy and an immunomodulatory compound for treatment of lymphoma
IL307598A (en) 2021-04-16 2023-12-01 Juno Therapeutics Inc T cell therapy in patients who have had prior stem cell transplant
CA3217914A1 (en) 2021-05-04 2022-11-10 Kevin BRAY Chimeric antigen receptors with mage-a4 specificity and uses thereof
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
JP2024517956A (en) 2021-05-14 2024-04-23 カイト ファーマ インコーポレイテッド Chimeric antigen receptor T cell therapy
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
AU2022324040A1 (en) 2021-08-04 2024-02-22 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
CN118043450A (en) 2021-08-24 2024-05-14 洛桑联邦理工学院 IL-10 expressing cells for enhancing cancer immunotherapy
WO2023081715A1 (en) 2021-11-03 2023-05-11 Viracta Therapeutics, Inc. Combination of car t-cell therapy with btk inhibitors and methods of use thereof
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
TW202342757A (en) 2021-12-17 2023-11-01 美商薩那生物科技公司 Modified paramyxoviridae attachment glycoproteins
TW202342498A (en) 2021-12-17 2023-11-01 美商薩那生物科技公司 Modified paramyxoviridae fusion glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150181A1 (en) 2022-02-01 2023-08-10 President And Fellows Of Harvard College Methods and compositions for treating cancer
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023159001A1 (en) 2022-02-15 2023-08-24 Kite Pharma, Inc. Predicting adverse events from immunotherapy
WO2023170606A1 (en) 2022-03-08 2023-09-14 Alentis Therapeutics Ag Use of anti-claudin-1 antibodies to increase t cell availability
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
WO2024030441A1 (en) 2022-08-02 2024-02-08 National University Corporation Hokkaido University Methods of improving cellular therapy with organelle complexes
US20240091261A1 (en) 2022-08-26 2024-03-21 Kite Pharma, Inc. Immune cell function
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells
US20240165160A1 (en) 2022-10-28 2024-05-23 Kite Pharma, Inc. Efficacy and durable response of immunotherapy
US20240158869A1 (en) 2022-10-28 2024-05-16 Kite Pharma, Inc. Factors for optimizing immunotherapy
WO2024091669A1 (en) 2022-10-28 2024-05-02 Ginkgo Bioworks, Inc. Chimeric antigen receptors comprising an intracellular domain pair
WO2024100604A1 (en) 2022-11-09 2024-05-16 Juno Therapeutics Gmbh Methods for manufacturing engineered immune cells

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US6040177A (en) 1994-08-31 2000-03-21 Fred Hutchinson Cancer Research Center High efficiency transduction of T lymphocytes using rapid expansion methods ("REM")
US20030170238A1 (en) 2002-03-07 2003-09-11 Gruenberg Micheal L. Re-activated T-cells for adoptive immunotherapy
WO2006060878A1 (en) 2004-12-10 2006-06-15 Peter Maccallum Cancer Institute Methods and compositions for adoptive immunotherapy
US20080131415A1 (en) 2006-11-30 2008-06-05 Riddell Stanley R Adoptive transfer of cd8 + t cell clones derived from central memory cells
WO2010025177A1 (en) 2008-08-26 2010-03-04 City Of Hope Method and compositions for enhanced anti-tumor effector functioning of t cells

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA981478B (en) 1997-02-21 1998-08-24 Vlaams Interuniv Inst Biotech Use of interleukin-15
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
EP1334188B1 (en) 2000-11-07 2006-08-30 City of Hope Cd19-specific redirected immune cells
US7070995B2 (en) 2001-04-11 2006-07-04 City Of Hope CE7-specific redirected immune cells
US20090257994A1 (en) 2001-04-30 2009-10-15 City Of Hope Chimeric immunoreceptor useful in treating human cancers
US7514537B2 (en) 2001-04-30 2009-04-07 City Of Hope Chimeric immunoreceptor useful in treating human gliomas
US7553494B2 (en) * 2001-08-24 2009-06-30 Corixa Corporation WT1 fusion proteins
US20030134415A1 (en) 2001-09-19 2003-07-17 Gruenberg Micheal L. Th1 cell adoptive immunotherapy
SI1806358T1 (en) * 2005-09-05 2010-06-30 Immatics Biotechnologies Gmbh Tumor-associated peptides binding promiscuously to human leukocyte antigen (HLA) class II molecules
EP1795599A1 (en) * 2005-12-09 2007-06-13 Schuler, Gerold, Prof. Dr. Methods for generating antigen-specific effector T cells
TW200848431A (en) * 2007-06-12 2008-12-16 Trubion Pharmaceuticals Inc Single-chain multivalent binding proteins with effector function
NZ743310A (en) * 2011-03-23 2022-11-25 Fred Hutchinson Cancer Center Method and compositions for cellular immunotherapy
JP5372297B1 (en) 2012-12-20 2013-12-18 三菱電機株式会社 In-vehicle device and program
US9108442B2 (en) 2013-08-20 2015-08-18 Ricoh Company, Ltd. Image forming apparatus

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US6040177A (en) 1994-08-31 2000-03-21 Fred Hutchinson Cancer Research Center High efficiency transduction of T lymphocytes using rapid expansion methods ("REM")
US20030170238A1 (en) 2002-03-07 2003-09-11 Gruenberg Micheal L. Re-activated T-cells for adoptive immunotherapy
WO2006060878A1 (en) 2004-12-10 2006-06-15 Peter Maccallum Cancer Institute Methods and compositions for adoptive immunotherapy
US20080131415A1 (en) 2006-11-30 2008-06-05 Riddell Stanley R Adoptive transfer of cd8 + t cell clones derived from central memory cells
WO2010025177A1 (en) 2008-08-26 2010-03-04 City Of Hope Method and compositions for enhanced anti-tumor effector functioning of t cells

Non-Patent Citations (56)

* Cited by examiner, † Cited by third party
Title
Abdulahad et al., "Persistent expansion of CD4+ effector memory T cells in Wegener's granulomatosis," Kidney International 70:938-947, 2006.
Altenschmidt et al., "Adoptive Transfer of in Vitro-Targeted, Activated T Lymphocytes Results in Total Tumor Regression,"The Journal of Immunology 159: 5509-5515, 1997.
Altenschmidt et al., "Cytolysis of Tumor Cells Expressing the Neu/erbB-2, erbB-3, and erbB-4 Receptors by Genetically Targeted Naive T Lymphocytes," Clinical Cancer Research 2: 1001-1008, Jun. 1996.
Baskar et al., "Unique Cell Surface Expression of Receptor Tyrosine Kinase ROR1 in Human B-Cell Chronic Lymphocytic Leukemia," Clin. Cancer Res. 14(2): 396-404, Jan. 15, 2008.
Berger et al., "Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates," The Journal of Clinical Investigation 118(1): 294-305, Jan. 2008.
Berger et al., "Adoptive transfer of virus-specific and tumor-specific T cell immunity," Current Opinion in Immunology 21: 224-232, 2009.
Blattman et al., "Cancer Immunotherapy: A Treatment for the Masses," Science 305: 200-205, Jul. 9, 2004.
Bleakley et al., "Molecules and mechanisms of the graft-versus-leukaemia effect," Nature Reviews Cancer 4: 371-380, May 2004.
Bollard et al., "Cytotoxic T Lymphocyte Therapy for Epstein-Barr Virus+ Hodgkin's Disease," J. Exp. Med. 200(12): 1623-1633, Dec. 20, 2004.
Brentjens et al., "Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15," Nature Medicine 9(3): 279-286, Mar. 2003.
Brentjens, R. et al., Clin Cancer Res., vol. 13 (2007), pp. 5426-5435. *
Butcher et al., "Lymphocyte homing and homeostasis," Science 272(5258): 60, 1996, 13 pages.
Chang et al., "Transgene-enforced co-stimulation of CD4+ T cells leads to enhanced and sustained anti-tumor effector functioning," Cytotherapy 9(8):771-784 (2007).
Cheadle et al., "Natural Expression of the CD19 Antigen Impacts the Long-Term Engraftment but Not Antitumor Activity of CD19-Specific Engineered T Cells," The Journal of Immunology 184(4): 1885-1896, Jan. 20, 2010.
Cheever et al., "Specificity of adoptive chemoimmunotherapy of established syngeneic tumors," The Journal of Immunology 125(2): 711-714, Aug. 1980.
Dotti et al., "Fifteen Years of Gene Therapy Based on Chimeric Antigen Receptors: Are We Nearly there Yet?" Human Gene Therapy 20:1229-1239 (Nov. 2009).
Dudley et al., "A phase I Study of Nonmyeloablative Chemotherapy and Adoptive Transfer of Autologous Tumor Antigen-Specific T Lymphocytes in Patients With Metastatic Melanoma," J. Immunother. 25(3): 243-251, 2002.
Dudley et al., "Adoptive Cell Transfer Therapy Following Non-Myeloablative but Lymphodepleting Chemotherapy for the Treatment of Patients With Refractory Metastatic Melanoma," J. Clin. Oncol. 23(10): 2346-2357, Apr. 1, 2005.
Dudley et al., "Adoptive Transfer of Cloned Melanoma-Reactive T Lymphocytes for the Treatment of Patients with Metastatic Melanoma," Journal of Immunotherapy 24(4): 363-373, 2001.
Dudley et al., "Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes," Science 298(5594): 850-854, Oct. 25, 2002.
Fukuda et al., "Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a," Proc. Natl. Acad. Sci. U S A 105(8): 3047-3052, Feb. 26, 2008.
Gattinoni et al., "Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells," The Journal of Clinical Investigation 115(6): 1616-1626, Jun. 2005.
Gattinoni et al., "Adoptive immunotherapy for cancer: building on success," Nat. Rev. Immunol. 6(5): 383-393, May 2006.
Hinrichs et al., "Adoptively transferred effector cells derived from naïve rather than central memory CD8+ T cells mediate superior antitumor immunity," PNAS 106(41): 17469-17474, Oct. 13, 2009.
Hinrichs et al., "Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy," Blood 117(3): 808-814, 2011.
Huang et al., "Sleeping Beauty Transposon-mediated Engineering of Human Primary T Cells for Therapy of CD19+ Lymphoid Malignancies," Molecular Therapy 16(3):580-589 (Mar. 2008).
Hudecek et al., "Naïve CD4+ T Cells Modified to Express a ROR1-Specific CAR Mediate Anti-Tumor Activity and Provide Superior Help to CD8+ ROR1-CART Cells," Blood (ASH Annual Meeting Abstracts) 118: Abstract 643, 2011, 3 pages.
Hudecek et al., "The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor," Blood 116(22): 4532-4541, 2010.
Isolation of CD4+ CD62L+ T cells, Miltenyi Biotec (4 pages) (2007).
Kershaw et al., "Gene-Engineered T Cells as a Superior Adjuvant Therapy for Metastatic Cancer," The Journal of Immunology 173: 2143-2150, 2004.
Kessels et al., "Immunotherapy through TCR gene transfer," Nature Immunology 2(10): 957-961, Oct. 2001.
Klein et al., "Gene Expression Profiling of B Cell Chronic Lymphocytic Leukemia Reveals a Homogeneous Phenotype Related to Memory B Cells," J. Exp. Med. 194(11): 1625-1638, Dec. 3, 2001.
Lamers et al., "Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells," Blood 117(1):72-82 (Jan. 6, 2011).
Lapalombella et al., "Lenalidomide treatment promotes CD154 expression on CLL cells and enhances production of antibodiesby normal B cells through a PI3-kinase-dependent pathway," Blood 115(13): 2619-2629, Apr. 1, 2010.
Mitsuyasu et al., "Prolonged survival and tissue trafficking following adoptive transfer of CD4ζ gene-modified autologous CD4+ and CD8+ T cells in human immunodeficiency virus-infected subjects," Blood 96(3): 785-793, Aug. 2000.
Moeller et al., "Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection," Blood 106: 2995-3003, 2005.
Morgan et al., "Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes," Science 314(5796): 126-129, Oct. 6, 2006.
Pahl-Seibert et al., "Highly Protective On Vivo Function of Cytomegalovirus IE1 Epitope-Specific Memory CD8 T Cells Purified by T-Cell Receptor-Based Cell Sorting," Journal of Virology 79(9): 5400-5413, May 2005.
Pule et al., "Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma," Nat. Med. 14(11):1264-1270, 2008. (17 pages).
Riddell et al., "Restoration of Viral Immunity in Immunodeficient Humans by the Adoptive Transfer of T Cell Clones," Science 257: 238-241, Jul. 10, 1992.
Riddell et al., "The use of anti-CD3 and anti-CD28 monoclonal antibodies to clone and expand human antigen-specific T cells," Journal of Immunological Methods 128: 189-201, 1990.
Rooney et al., "Infusion of Cytotoxic T Cells for the Prevention and Treatment of Epstein-Barr Virus-Induced Lymphoma in Allogeneic Transplant Recipients," Blood 92(5): 1549-1555, Sep. 1, 1998.
Rosenwald et al., "Relation of Gene Expression Phenotype to Immunoglobulin Mutation Genotype in B Cell Chronic Lymphocytic Leukemia," The Journal of Experimental Medicine 194(11): 1639-1647, Dec. 3, 2001.
Sallusto et al., "Central Memory and Effector Memory T Cell Subsets: Function, Generation, and Maintenance," Annu. Rev. Immunol. 22: 745-763, 2004.
Schmitt et al., "Maintenance of T Cell Specification and Differentiation Requires Recurrent Notch Receptor-Ligand Interactions," J. Exp. Med. 200(4): 469-479, Aug. 16, 2004.
Singh et al., "Redirecting Specificity of T-Cell Populations for CD19 Using the Sleeping Beauty System," Cancer Res. 68(8):2961-2971, 2008.
Singh et al., "Selective Reprogramming of CD 19-Specific T Cells with IL-21 and CD28 Signaling for Adoptive Immunotherapy of Acute Lymphoblastic Leukemia," Biology of Blood and Marrow Transplantation 15(2): 61-62, Feb. 1, 2009, Abstract No. 164, 2 pages.
Stanislawski et al., "Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer," Nature Immunology 2(10): 962-970, Oct. 2001.
Teng et al.,"Immunotherapy of Cancer Using Systemically Delivered Gene-Modified Human T Lumphocytes," Human Gene Therapy 15: 699-708, Jul. 2004.
Walker et al., "Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection," Blood 96: 467-474, 2000.
Walter et al., "Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor," N. Engl. J. Med. 333: 1038-1044, Oct. 19, 1995.
Wang et al., "Engraftment of human central memory-derived effector CD8+ T cells in immunodeficient mice," Blood 117(6): 1888-1898, 2011.
Wang et al., Cellular Immunotherapy for Follicular Lymphoma Using Genetically Modified CD20-Specific CD8+ Cytotoxic T Lymphocytes, Molecular Therapy 9(4): 577-586, Apr. 2004.
Westwood et al., "Genetic redirection of T cells for cancer therapy," Journal of Leukocyte Biology 87: 791-803, May 2010.
Xie et al., "Naive tumor-specific CD4+ T cells differentiated in vivo eradicate established melanoma," The Journal of Experimental Medicine 207(3):651-667 (Feb. 15, 2010).
Yee et al., "Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred T cells," Proc. Natl. Acad. Sci. USA 99(25): 16168-16173, Dec. 10, 2002.

Also Published As

Publication number Publication date
SG10201602253SA (en) 2016-05-30
BR112013024395A2 (en) 2016-09-20
NZ726162A (en) 2020-02-28
CN103502438A (en) 2014-01-08
AU2016238963B2 (en) 2018-05-17
US20180296602A1 (en) 2018-10-18
SG193591A1 (en) 2013-10-30
JP2016183194A (en) 2016-10-20
KR20140023931A (en) 2014-02-27
ZA201901167B (en) 2020-10-28
JP6203705B2 (en) 2017-09-27
AU2016238963A1 (en) 2016-11-03
CN106074601A (en) 2016-11-09
JP2021046453A (en) 2021-03-25
IL228603A0 (en) 2013-12-31
RU2013147157A (en) 2015-04-27
MX2013010793A (en) 2014-03-21
AU2018204208B2 (en) 2020-07-30
JP2023038386A (en) 2023-03-16
NZ743310A (en) 2022-11-25
US20140314795A1 (en) 2014-10-23
MX2018011917A (en) 2021-11-11
JP2014510108A (en) 2014-04-24
KR101976882B1 (en) 2019-05-09
BR112013024395B1 (en) 2021-10-26
EP2689010B1 (en) 2020-11-18
MX359513B (en) 2018-10-01
RU2688185C2 (en) 2019-05-21
AU2012230780B2 (en) 2016-10-27
CA2830953A1 (en) 2012-09-27
EP2689010A1 (en) 2014-01-29
JP2019108403A (en) 2019-07-04
CN110200997A (en) 2019-09-06
US9987308B2 (en) 2018-06-05
JP6877905B2 (en) 2021-05-26
CA2830953C (en) 2023-02-28
ES2841983T3 (en) 2021-07-12
WO2012129514A1 (en) 2012-09-27
AU2012230780A1 (en) 2013-05-02
AU2018204208A1 (en) 2018-07-05
IL228603B (en) 2021-03-25

Similar Documents

Publication Publication Date Title
US11065278B2 (en) Method and compositions for cellular immunotherapy
US10968431B2 (en) Adoptive transfer of CD8+ T cell clones derived from central memory cells
US20170246279A1 (en) Compositions and methods for boosting the efficacy of adoptive cellular immunotherapy
US20210137983A1 (en) Nk cell expansion and uses thereof
US11788072B2 (en) Activation of APC in immunotherapy
US20230019849A1 (en) Method for Preparing CD7-Negative, CD3-Positive T Cells
NZ714574A (en) Method and compositions for cellular immunotherapy
NZ714574B2 (en) Method and compositions for cellular immunotherapy
NZ726162B2 (en) Method and compositions for cellular immunotherapy
JP2022531814A (en) Amplification of modified cells and their applications

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT RECEIVED

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: FRED HUTCHINSON CANCER CENTER, WASHINGTON

Free format text: MERGER AND CHANGE OF NAME;ASSIGNORS:FRED HUTCHINSON CANCER RESEARCH CENTER;SEATTLE CANCER CARE ALLIANCE;REEL/FRAME:060329/0793

Effective date: 20220330

AS Assignment

Owner name: FRED HUTCHINSON CANCER CENTER, WASHINGTON

Free format text: MERGER AND CHANGE OF NAME;ASSIGNORS:FRED HUTCHINSON CANCER RESEARCH CENTER;SEATTLE CANCER CARE ALLIANCE;REEL/FRAME:060722/0441

Effective date: 20220330